Next Article in Journal
Pharmacology and Rationale for Seralutinib in the Treatment of Pulmonary Arterial Hypertension
Next Article in Special Issue
Blood Proteome Profiling Reveals Biomarkers and Pathway Alterations in Fragile X PM at Risk for Developing FXTAS
Previous Article in Journal
Insights into the Inhibitory Mechanisms of the Covalent Drugs for DNMT3A
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Long COVID as a Tauopathy: Of “Brain Fog” and “Fusogen Storms”

by
Adonis Sfera
1,2,3,*,
Leah Rahman
4,
Carlos Manuel Zapata-Martín del Campo
5 and
Zisis Kozlakidis
6
1
Paton State Hospital, 3102 Highland Ave, Patton, CA 92369, USA
2
School of Behavioral Health, Loma Linda University, 11139 Anderson St., Loma Linda, CA 92350, USA
3
Department of Psychiatry, University of California, Riverside 900 University Ave, Riverside, CA 92521, USA
4
Department of Neuroscience, University of Oregon, 222 Huestis Hall, Eugene, OR 97401, USA
5
Instituto National de Cardiologia, Juan Badiano 1, Belisario Domínguez Secc 16, Tlalpan, Ciudad de México 14080, Mexico
6
International Agency for Research on Cancer, World Health Organization, 69000 Lyon, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(16), 12648; https://doi.org/10.3390/ijms241612648
Submission received: 13 July 2023 / Revised: 4 August 2023 / Accepted: 6 August 2023 / Published: 10 August 2023
(This article belongs to the Special Issue Genomics in Neurodegenerative Diseases)

Abstract

:
Long COVID, also called post-acute sequelae of SARS-CoV-2, is characterized by a multitude of lingering symptoms, including impaired cognition, that can last for many months. This symptom, often called “brain fog”, affects the life quality of numerous individuals, increasing medical complications as well as healthcare expenditures. The etiopathogenesis of SARS-CoV-2-induced cognitive deficit is unclear, but the most likely cause is chronic inflammation maintained by a viral remnant thriving in select body reservoirs. These viral sanctuaries are likely comprised of fused, senescent cells, including microglia and astrocytes, that the pathogen can convert into neurotoxic phenotypes. Moreover, as the enteric nervous system contains neurons and glia, the virus likely lingers in the gastrointestinal tract as well, accounting for the intestinal symptoms of long COVID. Fusogens are proteins that can overcome the repulsive forces between cell membranes, allowing the virus to coalesce with host cells and enter the cytoplasm. In the intracellular compartment, the pathogen hijacks the actin cytoskeleton, fusing host cells with each other and engendering pathological syncytia. Cell–cell fusion enables the virus to infect the healthy neighboring cells. We surmise that syncytia formation drives cognitive impairment by facilitating the “seeding” of hyperphosphorylated Tau, documented in COVID-19. In our previous work, we hypothesized that the SARS-CoV-2 virus induces premature endothelial senescence, increasing the permeability of the intestinal and blood–brain barrier. This enables the migration of gastrointestinal tract microbes and/or their components into the host circulation, eventually reaching the brain where they may induce cognitive dysfunction. For example, translocated lipopolysaccharides or microbial DNA can induce Tau hyperphosphorylation, likely accounting for memory problems. In this perspective article, we examine the pathogenetic mechanisms and potential biomarkers of long COVID, including microbial cell-free DNA, interleukin 22, and phosphorylated Tau, as well as the beneficial effect of transcutaneous vagal nerve stimulation.

1. Introduction

The COVID-19 pandemic may be succeeded by prolonged sequelae that could affect the life quality of many patients while at the same time increasing medical complications and healthcare expenditures. Cognitive impairment, one of the lingering symptoms, often referred to by patients as “brain fog”, remains poorly defined; however, viral presence in reservoirs and the “seeding” of pathological Tau may account for cognitive difficulties.
Tauopathies are neurodegenerative disorders marked by the accumulation of neurofibrillary tangles and paired helical filaments in neurons and glia, leading to apoptosis. Tauopathies can be primary (Tau is the main disease driver) or secondary (Tau aggregation is triggered by a different insult). The most common secondary tauopathies—frontotemporal dementia (FTD) and Alzheimer’s disease (AD)—comprise the largest share of late-life neurodegenerative disorders, linking pTau to this pathology [1]. Tau is encoded by the microtubule-associated protein tau (MAPT) gene and has been implicated in the homeostasis of axonal and synaptic transmission [2]. Several tauopathies, including subacute sclerosing panencephalitis and postencephalitic parkinsonism, have been associated with viral infections, suggesting that long COVID may be driven by a similar mechanism [3,4,5].
Long COVID refers to the clinical manifestations that develop during or after COVID-19 infection and continue for at least 12 weeks [6]. Due to the absence of specific tests for this syndrome, long COVID is currently diagnosed by history taking and physical examination. For this reason, it is difficult to differentiate long COVID from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reinfection or the activation of dormant viruses, such as Herpes virus (HSV) [7]. By the same token, COVID-associated neurocognitive dysfunction (CAND) cannot be readily parsed out from other cognitive dysfunctions, whether new or pre-existing [8].
To effectively infect an organism, enveloped viruses must first usurp the plasma membrane fusogens and coalesce with host cells. Upon successful entry into the cytosol, the SARS-CoV-2 virus proceeds to fuse host cells with each other, forming large, multinucleated syncytial structures. These formations enhance infectivity by allowing infection as well as hyperphosphorylated Tau (pTau—a newly identified marker of long COVID—to spread “from within”, increasing the efficacy of dissemination. For example, the fusion of infected with noninfected cells proliferates the contagion more rapidly than invading cells one by one [9,10,11,12]. Furthermore, lipopolysaccharide (LPS) was demonstrated to enhance the detrimental effects of pTau, linking this gut-originating endotoxin to tauopathies [13].
In a previous article, we connected the SARS-CoV-2 virus with premature endothelial senescence, gut-barrier dysfunction, and the subsequent translocation of microbes and/or their molecules into host tissues and organs, including the brain [14]. Gut microbes are immunologically tolerated in the gastrointestinal (GI) tract, however, depending on the microorganism, they may trigger vehement immunogenicity upon migration across the lamina propria [15]. Indeed, novel studies have identified microbial DNA, and LPS in the peripheral blood of COVID-19 patients, suggesting that this virus facilitates translocation [16,17,18,19].
Novel studies have shown that the S protein of the SARS-CoV-2 virus binds directly to LPS, facilitating contagion as well as inflammation [20,21]. In addition, the S antigen-associated fusogen—Ca2+/calmodulin-dependent protein kinase II (CaMKII)—implicated in cognitive dysfunction promotes cell–cell fusion, augmenting the virulence of SARS-CoV-2 [22,23]. Moreover, viral attachment to angiotensin-converting enzyme-2 (ACE-2), upregulates angiotensin II (ANG II), contributing to both Tau hyperphosphorylation and dysfunctional acetylcholine (ACh) signaling [14,24,25] (Figure 1).
Mitochondria, crucial for host innate immunity, are exploited by many viruses, including SARS-CoV-2, to avert detection and ensure proper replication in host cells. For example, SARS-CoV-2 antigens (nonstructural protein 4 and 9 (NSP4, NSP9) and open reading frame 9C (ORF9C) disrupt the organelle, generating mitochondrial reactive oxygen species (mROS), which promote the development of pTau [9,26,27,28]. In this regard, assays of phosphorylated Tau at threonine 217 (pTau-217) and threonine 181 (pTau-181), developed by Lilly and Simoa®®, are being utilized as tauopathy blood markers, respectively. We suggest that the presence of these biomolecules can also accurately diagnose long COVID (Table 1).
Novel studies have shown that premature cellular senescence promotes pTau and upregulates intracellular calcium (Ca2+), activating the fusion machinery [29,30,31,32] (Figure 2). For example, Ca2+, released from the endoplasmic reticulum (ER) and/or imported from the extracellular compartment activates TMEM16F, a lipid scramblase that triggers cell–cell fusion by externalizing phosphatidylserine (ePS) [33] (Figure 2). In addition, premature cellular senescence upregulates intracellular iron, a biometal indispensable for viral proliferation that may increase oxidative stress and telomere erosion, causing further senescence.
The infection with the SARS-CoV-2 virus has been shown to activate human endogenous retroviruses (HERVs), a large source of ancestral fusogens likely implicated in long COVID [34,35]. HERVs are embedded in the human DNA and, with few exceptions, are epigenetically suppressed. However, under pathological circumstances, these viral fossils can be expressed, causing pathology, including cancer and neurodegenerative disorders [36].
In this perspective article, we take a closer look at long COVID as a probable tauopathy, discussing potential biomarkers such as microbial cell-free DNA (mcfDNA), interleukin 22 (IL-22), and phosphorylated Tau (pTau).
We also review the beneficial effects of noninvasive vagus nerve stimulation as a therapeutic intervention for long COVID.
Table 1. Potential markers for microbial translocation and tauopathy.
Table 1. Potential markers for microbial translocation and tauopathy.
Marker TypeMarkerAssayReferences
Gut barrierIL-22Singulex-Erenna®®[37]
Gut barriermcfDNAKarius Test®®[38]
NeurodegenerationpTau 217Lilly[39]
NeurodegenerationpTau 181Simoa®®[40]
NLRCICEntotic cell death assays[41]
Example: downregulated IL-22 with upregulated mcfDN, pTau, and NLR would reflect microbial translocation and the emergence of tauopathy. (Table 1).

2. SARS-CoV-2 Reservoirs

The symptoms of long COVID are likely caused by a viral remnant in select body reservoirs where the pathogen is shielded from host defenses. For example, at 15 months post-infection follow-up, the S protein of the SARS-CoV-2 virus was found in monocytes, indicating that these cells could comprise viral reservoirs [42]. Other studies have found that intestinal epithelial cells (IECs), adipocytes, and endothelial cells (ECs), as well as astrocytes and microglia, could serve as potential SARS-CoV-2 reservoirs [43,44,45,46]. In sanctuary cells, the virus has been found to associate with ferritin and pTau, linking this pathogen to protein misfolding and iron dyshomeostasis [47,48]. Interestingly, monocytes, microglia, and macrophages are known human immunodeficiency virus (HIV) reservoirs, suggesting that they could also harbor the SARS-CoV-2 virus [49].
At the molecular level, the AhR/STAT3/IL-22 axis exhibits antiviral and anti-inflammatory properties as it connects the cholinergic anti-inflammatory pathway (CAP) with gut microbes. In this regard, transcutaneous auricular vagus nerve stimulation (taVNS), a modality that increases the antiviral neurotransmitter ACh, may improve both cognition and the gut-barrier function [50,51] (Figure 1). Conversely, the aberrant phosphorylation of the AhR/STAT3/IL-22 axis can exacerbate viral infection and microbial translocation [52,53].
In the CNS, long COVID was associated with gray matter loss, likely accounting for the impaired attention, concentration, and executive function documented in this condition [44,54,55,56]. Several studies have linked long COVID to the reactive astrocytes, which the virus may enter via a basigin (BSG) receptor, a protein implicated in microvascular disease [57,58]. This is significant as astrocytes, established HIV reservoirs, are essential for the neuro-vascular unit and may easily capture the SARS-CoV-2 virus from the cerebral circulation [59,60,61]. In addition, as microglia are known HIV reservoirs, these cells may promote long COVID by also housing the SARS-CoV-2 virus [62]. Indeed, both astrocytes and microglia can assume neurotoxic phenotypes, eliminating healthy neurons and synapses—a process that could account for “brain fog” [44].
The likely existence of SARS-CoV-2 viral reservoirs highlights the unmet need of eradicating this as well as the HIV virus from the sanctuary cells. This brings CAP to the forefront of long COVID treatment. Indeed, at present, no α7nAChR agonist has been approved (and nicotine is a nonspecific contributor), and VN stimulation (VNS) may be the only available modality for CAP augmentation and the removal of viruses from reservoirs [63,64]. The activation of α7nAChR in persons living with HIV has ameliorated HIV-associated neurocognitive disorders (HAND), suggesting that this modality can clear viral reservoirs, areas where the highly active antiretroviral therapy (HAART) cannot access [65]. We hypothesize that taVNS can clear both SARS-CoV-2 and HIV from viral reservoirs by activating AhR/STAT3/IL-22.

3. SARS-CoV-2 Virus-Induced Cell–Cell Fusion

Enveloped viruses, including SARS-CoV-2, are known for inducing premature cellular senescence, a program of permanent replication arrest with an active metabolism, generating a unique secretome known as senescence-associated secretory phenotype (SASP). They upregulate intracellular iron, and Ca2+, senescent and fused cells are hospitable to viruses that require these elements to replicate [66,67,68]. Fused cells, characterized by ePS, promote immunosuppression, enabling the virus to avert host immunity [69]. However, cellular senescence-upregulated intracellular iron may trigger ferroptosis, a nonapoptotic cell death caused by the oxidation of endoplasmic reticulum (ER) lipids in the absence of antioxidants. Indeed, due to SARS-CoV-2’s affinity for astrocytes, the virus likely disrupts the generation of glutathione peroxidase 4 (GPX4), a selenocysteine, which is in charge of repairing oxidized phospholipids. GPX4 is synthesized by astrocytes and shuttled to the neurons, where it prevents lipid peroxidation and ferroptosis [70].
In contrast to ferroptosis, which involves neuronal death, ferrosenescence is a phenotype in which the cells remain alive but iron-damaged genomes enable the mobilization of transposable elements (TEs) known colloquially as” jumping genes”. TEs are DNA segments that can extricate themselves from the genome and reinsert in the double helix at a different location. TE mobilization has been associated with “fusogen storms”—the excessive release of fusion molecules from HERVs (viruses acting as TEs) [71]. Moreover, pTau was demonstrated to further mobilize TEs, enhancing fusogen storms while generating a hospitable milieu for viral progeny [72].
It is unclear at this time whether fused neurons are viable in humans and for what length of time. A recent study in Caenorhabditis elegans (C. elegans) found that an aberrant expression of fusogens in neurons alters the connectome as well as the behavior, although sensation remains intact [25]. It is difficult to extrapolate these data to humans; however, connectivity was demonstrated to affect cognition [73].
In a previous article, we defined ferrosenescence as “neurodegeneration-by-iron”, that is, premature molecular aging due to iron-induced damage to both DNA and the genomic repair systems, especially the p53 [74]. Since p53 also drives natural killer cells (NKCs), which under normal circumstances remove ferrosenescent cells, dysfunctional p53 may lead to iron deposition in the central nervous system (CNS) [75]. Indeed, excessive iron in the caudate and putamen was documented in long COVID, linking this condition to ferrosenescence [76,77].
The SARS-CoV-2 virus can alter iron metabolism by several mechanisms. For example, the S1 protein/ACE-2 attachment upregulates ANG II, increasing intracellular iron via angiotensin II type 1 receptors (AT-1Rs) (Figure 3). The S2 antigen of the SARS-CoV-2 virus disables p53, inducing ferrosenescence due to unrepaired DNA. The virus also attacks hemoglobin via ORF3 and ORF10 antigens, releasing more iron for viral replication [78,79]. On the other hand, sartans and iron chelators avert ferrosenescence by blocking AT-1Rs and lowering the excess iron (Figure 3).
Both physiological and pathological aging were associated with syncytia formation and excessive intracellular Ca2+ and iron [80,81]. Moreover, the SARS-CoV-2 virus disseminates pTau to healthy cells, both directly by piercing the cell membranes and indirectly via extracellular vesicles (EVs) [82,83].

4. Fusion of Postmitotic Cells: Heart and Brain Syncytia

Several body tissues are structured as anatomical or functional syncytia. For example, the cardiac muscle maintains distinct cellular boundaries while functioning as a syncytium [84]. In contrast, skeletal muscle fibers and placental villous trophoblasts form anatomical syncytia, generating giant structures with a common cytoplasm and multiple nuclei [85].
Several viruses are known for inducing pathological cell–cell fusion, which in the heart and skeletal muscle may be superimposed on physiological syncytia, causing pathology. Viral pathogens can also drive postmitotic cells such as cardiomyocytes and myocytes to re-enter mitosis, probably contributing to cardiomyopathy in the myocardium and fatigue, frailty, or weakness in skeletal muscle [86,87].
In the CNS, astrocytes and oligodendrocytes form syncytia as they communicate via connexin channels [88]. These structures may arise from postmitotic neurons re-entering the cell cycle and forming aneuploid or polyploid cells, which is a phenomenon reported more than two decades ago [29]. As opposed to the early studies that assumed that fused neurons always undergo apoptosis, new research has found that these cells can stay indefinitely in fused states and remain functional [89,90].
The idea of cell–cell fusion in the brain has been around since the 19th century when Golgi and Cajal had opposite views on whether neurons were distinct entities or formed syncytial structures [91]. The advent of electron microscopy in the middle of the 20th century revealed that the neurons were individual cells, while astrocytes formed functional syncytia [92].
Under pathological circumstances, including viral infections, neurons can fuse with each other or with glial cells, forming syncytia (Figure 4). For example, the fusion of Purkinje neurons with bone marrow cells was demonstrated in both humans and rodents, suggesting that neurons can fuse not only with each other but also with somatic cells [29,93]. In addition, multinucleated neurons in the supraoptic nucleus were found in patients with pneumonia, suggesting that fusogenic viruses can access the brain [94]. Moreover, neuronal syncytia were demonstrated in both normal and pathological aging, suggesting that aneuploidy and polyploidy could be more common in the human brain than currently believed [95,96]. Indeed, somatic mosaicism (genomic differences from neuron to neuron) was discovered with the help of a new technique capable of discerning chromosomes in individual cells [97]. The fact that neurons have different DNA from one cell to another is significant and may be the result of genome reorganization after viral infections or TE mobilization. This suggests that viruses may contribute to the variability of human brain function in both health and disease.
Taken together, cellular senescence and cell–cell fusion are physiological processes exploited by viruses to facilitate replication in human cells. Upregulated iron and Ca2+ in senescent cells generate a virus-friendly milieu while increasing the risk of host cognitive dysfunction.

5. SARS-CoV-2 Virus Activates Human Endogenous Retroviruses (HERVs)

Human endogenous retroviruses (HERVs) are viral relics that were integrated into the genome in ancient times and currently comprise about 8% of human DNA [98]. Most HERVs consist of damaged genes that cannot be transcribed and are suppressed by p53, which, aside from repairing the genome, blocks viruses from integrating into human DNA [99]. To overcome p53, cancers and viruses often exploit ubiquitination, an epigenetic mechanism that terminates the action of p53 [100].
Cell–cell fusion, a physiological mechanism of wound healing, is activated by cell membrane lesions, which drive the “wounded” cell to fuse with its healthy neighbors, preventing apoptosis by spillage of the intracellular content [101]. Viruses exploit wound healing by puncturing cell membranes to activate the fusion machinery [102]. To accomplish this task, the SARS-CoV-2 virus exploits arginine, a unique amino with side chains capable of piercing cell membranes [103,104]. For example, the S antigen of the SARS-CoV-2 virus contains a polybasic cleavage motif, “Proline-Arginine-Arginine-Alanine (PRRA)”, in which the bi-arginine forms a pore in the cell membrane, initiating fusion [105]. Aside from utilizing arginine, the SARS-CoV-2 virus also activates HERVs, providing additional fusogens—a “plan B”—for ensuring viral entry [34].
Over thousands of years, several HERVs have been “domesticated” and have assumed physiological functions in the human body. For example, HERV-W ENV encodes for a physiological placental fusogen, syncytin-1, which plays a key role in pregnancy by enabling the formation of syncytiotrophoblasts [106]. In addition, HERV-FRD, another “domesticated” endogenous retrovirus, encodes for syncytin-2, a fusogen that also plays a key role in placentation [107,108]. In the CNS, the neuronal gene Arc is a “domesticated” HERV and a master regulator of synaptic plasticity; it is derived from the ancestral Ty3/gypsy retrovirus, implicated in memory formation [109].
DNA damage can awaken suppressed HERVs, triggering pathology, including cancer and neuropsychiatric illness [110]. As p53 opposes the transcription of HERVs as well as the integration of exogenous viruses into human DNA, SARS-CoV-2 can release fusogens by blocking p53, disinhibiting the transcription of syncytin-1 and 2 [111] (Figure 5). In consequence, enhanced syncytia formation increases the systemic symptoms of long COVID, including “brain fog”, by several mechanisms, including HERV activation.
In the brain, excessive syncytin-1 can activate several pro-inflammatory and autoimmune cascades, triggering neuropsychiatric pathology, including multiple sclerosis (MS) [112]. Moreover, the SARS-CoV-2 inhibition of p53 destabilizes the genome, mobilizing TEs and facilitating the transcription of fusogens, including syncytins [113]. Along this line, impaired p53 predisposes to cancer, suggesting that SARS-CoV-2 may be an oncogenic virus [114,115,116]. Indeed, pTau was implicated in tumorigenesis, while HERVs spread pTau, connecting ancient viruses with present-day cancer [82]. In addition, since COVID-19 disrupts DNA repair, it may inhibit cell-cycle arrest genes, inducing postmitotic cells to engage in mitosis [117].

6. Microbial Translocation

It has been established that the SARS-CoV-2 virus employs several mechanisms to induce premature endothelial senescence and a dysfunctional intestinal and blood–brain barrier (BBB). This contributes to the translocation of microbes and/or their molecules from the GI tract into host tissues and organs, eventually reaching the brain (Figure 6).
Microbial translocation from the GI tract into the systemic circulation attracted the attention of clinicians and researchers during the human immunodeficiency virus 1 (HIV-1) epidemic in the 1980s. This virus has been known for depleting gut interleukin 22 (IL-22), leading to increased intestinal permeability and microbial dissemination in host tissues [118].
Various microbial translocation markers have been described since the 1980s, including LPS-binding protein (LBP), soluble CD14 (sCD14), and endotoxincore antibody (EndoCAb) [119]. We propose that IL-22 and microbial cell-free DNA (mcfDNA) comprise second-generation, high-quality markers of intestinal permeability and microbial migration. In addition, we believe that pTau 217 and pTau 181 can identify tauopathies as well as long COVID.
Neutrophil-to-lymphocyte ratio (NLR) is an inflammatory marker that may be elevated in long COVID due to lymphopenia. Low lymphocyte count may be caused by cell-in-cell (CIC) structures, which are phenomena described by Karl J. Eberth in the 19th century. CIC formations refer to cells internalized by other cells and are phenomena observed in cancer and lymphocytes-in-IECs documented in both COVID-19 and long COVID [120,121]. This may be significant as the internalized lymphocytes, likely infected with SARS-CoV-2, may comprise unique viral reservoirs, maintaining long COVID.

6.1. Interleukin 22 (IL-22)

Discovered in 2000, IL-22 is a member of the IL-10 family of cytokines generated by several lymphocyte types, including T helper (Th) 17 cells, γδ T cells, NKCs, and innate lymphoid cells (ILCs) [122]. IL-22 controls several IEC functions, including mucus formation, permeability, and the synthesis of complement and antimicrobial peptides (AMPs), indicating that this cytokine functions as the master regulator of gut-barrier permeability [123].
The crosstalk between IL-22 and its receptor (IL-22R), a dimeric protein comprised of IL-22R1 and IL-10R2, activates the JAK/STAT pathway, a key antibacterial and antiviral system that protects against gut pathogens [124,125]. IL-22 exerts antiviral properties against COVID-19 and influenza, as well as beneficial action in inflammatory bowel disease (IBD) [126,127,128,129]. Singulex-Erenna®®, a standardized IL-22 peripheral blood assay, should be studied further not only for IBD and viral infections but also for neuropsychiatric pathology [37].

6.2. Microbial Cell-Free DNA (mcfDNA)

mcfDNA is a noninvasive, high-throughput sequencing technology that detects pathogens in the peripheral blood [130]. Presently, mcfDNA has only been utilized in sepsis; however, given its high sensitivity and specificity, it likely comprises a valid biomarker of gut permeability [131]. For example, the Karius Test®®, developed in 2017, can detect mcfDNA in the peripheral blood, suggesting that it could be a diagnostic tool for the microorganisms associated with neuropsychiatric disorders [38].

6.3. Hyperphosphorylated Tau (pTau)

pTau, a central marker of tauopathy, has been detected in several peripheral organs, including the gut, in which it is associated with IBD [132,133]. Aside from the aggregated long fibrils, several studies found short pTau in COVID-19-associated peripheral neuropathy [134]. pTau 217 and pTau 181 have been patented by Lilly and Simoa®® for AD; however, they could also diagnose long COVID [39,40].

6.4. Neutrophil-to-Lymphocyte Ratio (NLR)

NLR is obtained from the peripheral blood and reflects the ratio between innate immunity (neutrophils) and the adaptive immune system (lymphocytes) [135]. Low lymphocyte counts, lasting for up to 16 weeks after the acute COVID-19 illness were detected in many patients, suggesting that lymphopenia could be a reliable biomarker of long COVID [136].
Lymphopenia may be caused by cell-in-cell (CIC) structures, which are phenomena referring to lymphocytes internalized in IECs [41,120,137]. CIC structures, commonly found in cancer, can be driven by mitosis and potentially tumor-induced autophagy [138]. Interestingly, lymphopenia has been associated with mitochondrial dysfunction [139]. In SARS-CoV-2 infections, mitochondria play an essential role as, aside from the interaction with SARS-CoV-2 NSPs, the virus can disrupt immune responses by altering the release of type I interferon (IFN-I) via mitochondria anti-viral signaling (MAVS) adaptor molecules. Indeed, viral proteins can activate caspases 8 and 9, promoting the release of cytochrome c, activating the mitochondrial death pathway. In addition, the SARS-CoV-2 virus can also cause gastrointestinal infections, altering the mitochondria–gut microbiota crosstalk, a key process for intestinal homeostasis [140].

7. Vagus Nerve Stimulation

In this section, we examine the physiological role of the vagus nerve and CAP in averting microbial translocation, highlighting the manipulation of cholinergic signaling as a treatment strategy for long COVID, including the “brain fog”.
The vagus nerve is composed of 20% efferent and 80% afferent fibers that support two-way communication between the brain and the body’s periphery. This is significant as it indicates that despite autonomy (ENS can manage the GI function after vagotomy), the gut is regulated centrally via the efferent vagus and neuro-immune pathways.
Novel studies have shown that the SARS-CoV-2 virus disrupts CAP, altering cholinergic signaling and the gut–brain axis [141]. In this regard, SARS-CoV-2 may exacerbate myasthenia gravis, emphasizing interference with neuronal and non-neuronal ACh [142,143].
Novel studies have found that α7nAChR dysfunction likely precedes Tau pathology, emphasizing the importance of cholinergic signaling in preventing tauopathies [144]. On the other hand, noninvasive taVNS may restore not only the integrity of the gut barrier but also “drain” viral reservoirs in microglia, monocytes, ECs, or IECs, eradicating the latent infection.
Currently, migraine headaches and some seizures are being treated with transcutaneous vagal nerve stimulation (tVNS), a well-tolerated modality that is easy to administer. A tVNS variant, spleen ultrasound neuromodulation, is based on the recent finding that memory T cells generate ACh in the gut and spleen, indicating that this modality could restore the homeostasis of adaptive immunity [145,146,147,148].
Taken together, the vagus nerve regulates ENS via CAP. At the molecular level, CAP influences intestinal permeability and gut microbiota composition via the AhR/STAT3/IL-22 axis. Under pathological conditions, this axis can be impaired, disrupting the gut–brain communication—a deficit that is restored by taVNS.
Recent studies have found that the SARS-CoV-2 virus can hijack the signal transducer and activator of transcription 1 (STAT1), hyperactivating STAT3 and disrupting the AhR/STAT3/IL-22 axis [149,150]. It was hypothesized that activated STAT3 maintains the symptoms of long COVID by altering immunity, a condition reminiscent of the rare genetic disorder STAT3 gain of function (GOF) [151,152,153]. Along this line, several studies have shown that α7nAChR stimulation inactivates STAT3, emphasizing the mechanism by which taVNS may ameliorate long COVID [154,155,156] (Figure 7). Indeed, taVNS lowers endothelial senescence, deactivates macrophages/monocytes/microglia, and restores gut-barrier homeostasis.

8. Conclusions

Long COVID is the proof of concept that the SARS-CoV-2 virus can linger in viral reservoirs, including microglia, EC, or IECs, engendering long COVID.
The S antigen of the SARS-CoV-2 virus contains fusogens that pierce host plasma membranes, initiating cell–cell fusion. Fused, aneuploid, or polyploid neurons, encountered in normal aging as well as in major cognitive disorders, likely account for the “brain fog” experienced by many patients with long COVID. Moreover, SARS-CoV-2 and HERVs “seed” pTau, suggesting that long COVID shares many pathogenetic traits with tauopathies. Furthermore, excessive pTau disrupts CAP and ACh signaling, compromising the gut barrier and contributing to the translocation of LPS and microbes outside the GI tract. LPS enhances viral infectivity and at the same time augments the generation of pTau, further linking long COVID to tauopathies.
taVNS restores the integrity of CAP as well as the gut-barrier function. In addition, upregulated intracellular ACh, a neurotransmitter with antiviral properties, may eradicate the virus from reservoirs, indicating its likely usefulness for both long COVID and HIV.

Author Contributions

A.S.: conceptualization and writing; L.R.: data curation; C.M.Z.-M.d.C.: writing, review, and editing; Z.K.: editing, writing, and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Silva, M.C.; Haggarty, S.J. Tauopathies: Deciphering Disease Mechanisms to Develop Effective Therapies. Int. J. Mol. Sci. 2020, 21, 8948. [Google Scholar] [CrossRef]
  2. Venkatramani, A.; Panda, D. Regulation of neuronal microtubule dynamics by tau: Implications for tauopathies. Int. J. Biol. Macromol. 2019, 133, 473–483. [Google Scholar] [CrossRef] [PubMed]
  3. Pratt, J.; Lester, E.; Parker, R. Could SARS-CoV-2 cause tauopathy? Lancet Neurol. 2021, 20, 506. [Google Scholar] [CrossRef] [PubMed]
  4. Hamdy, N.M.; Shaker, F.H.; Zhan, X.; Basalious, E.B. Tangled quest of post-COVID-19 infection-caused neuropathology and what 3P nano-bio-medicine can solve? EPMA J. 2022, 13, 261–284. [Google Scholar] [CrossRef] [PubMed]
  5. Villa, C.; Rivellini, E.; Lavitrano, M.; Combi, R. Can SARS-CoV-2 Infection Exacerbate Alzheimer’s Disease? An Overview of Shared Risk Factors and Pathogenetic Mechanisms. J. Pers. Med. 2022, 12, 29. [Google Scholar] [CrossRef]
  6. Jarrott, B.; Head, R.; Pringle, K.G.; Lumbers, E.R.; Martin, J.H. “LONG COVID”-A hypothesis for understanding the biological basis and pharmacological treatment strategy. Pharmacol. Res. Perspect. 2022, 10, e00911. [Google Scholar] [CrossRef]
  7. Barton, E.S.; White, D.W.; Cathelyn, J.S.; Brett-McClellan, K.A.; Engle, M.; Diamond, M.S.; Miller, V.L.; Virgin, H.W., IV. Herpesvirus latency confers symbiotic protection from bacterial infection. Nature. Nature 2007, 447, 326–329. [Google Scholar] [CrossRef]
  8. Watson, A.M.M.; Prasad, K.M.; Klei, L.; Wood, J.A.; Yolken, R.H.; Bradford, L.D.; Calkins, M.E.; Richard, J.; Edwards, N.; Savage, R.M.; et al. Persistent infection with neurotropic herpes viruses and cognitive impairment. Psychol. Med. 2012, 43, 1023–1031. [Google Scholar] [CrossRef] [Green Version]
  9. Pal, D. Spike protein fusion loop controls SARS-CoV-2 fusogenicity and infectivity. J. Struct. Biol. 2021, 213, 107713. [Google Scholar] [CrossRef]
  10. Narjes, B.; Khalid, I. COVID-19 and Neurodegenerative Diseases: Prion-Like Spread and Long-Term Consequences. J. Alzheimer’s Dis. 2022, 88, 399–416. [Google Scholar]
  11. Reiken, S.; Sittenfeld, L.; Dridi, H.; Liu, Y.; Liu, X.; Marks, A.R. Alzheimer’s-like signaling in brains of COVID-19 patients. Alzheimer’s Dement. 2022, 18, 955–965. [Google Scholar] [CrossRef]
  12. Lukiw, W.J.; Jaber, V.R.; Pogue, A.I.; Zhao, Y. SARS-CoV-2 Invasion and Pathological Links to Prion Disease. Biomolecules 2022, 12, 1253. [Google Scholar] [CrossRef] [PubMed]
  13. Roe, A.D.; Staup, M.A.; Serrats, J.; Sawchenko, P.E.; Rissman, R.A. Lipopolysaccharide-induced tau phosphorylation and kinase activity--modulation, but not mediation, by corticotropin-releasing factor receptors. Eur. J. Neurosci. 2011, 34, 448–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sfera, A.; Osorio, C.; Jafri, N.; Diaz, E.L.; Campo Maldonado, J.E. Intoxication with Endogenous Angiotensin II: A COVID-19 Hypothesis. Front. Immunol. 2020, 11, 1472. [Google Scholar] [CrossRef] [PubMed]
  15. Fine, R.L.; Manfredo Vieira, S.; Gilmore, M.S.; Kriegel, M.A. Mechanisms and consequences of gut commensal translocation in chronic diseases. Gut Microbes 2019, 11, 217–230. [Google Scholar] [CrossRef] [Green Version]
  16. Oliva, A.; Miele, M.C.; Di Timoteo, F.; De Angelis, M.; Mauro, V.; Aronica, R.; Al Ismail, D.; Ceccarelli, G.; Pinacchio, C.; D’ettorre, G.; et al. Persistent Systemic Microbial Translocation and Intestinal Damage During Coronavirus Disease-19. Front. Immunol. 2021, 12, 708149. [Google Scholar] [CrossRef]
  17. Teixeira, P.C.; Dorneles, G.P.; Filho, P.C.S.; da Silva, I.M.; Schipper, L.L.; Postiga, I.A.; Neves, C.A.M.; Junior, L.C.R.; Peres, A.; de Souto, J.T.; et al. Increased LPS levels coexist with systemic inflammation and result in monocyte activation in severe COVID-19 patients. Int. Immunopharmacol. 2021, 100, 108125. [Google Scholar] [CrossRef]
  18. Tetz, V.; Tetz, G. Bacterial DNA induces the formation of heat-resistant disease-associated proteins in human plasma. Sci. Rep. 2019, 9, 17995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Dereschuk, K.; Apostol, L.; Ranjan, I.; Chakladar, J.; Li, W.T.; Rajasekaran, M.; Chang, E.Y.; Ongkeko, W.M. Identification of Lung and Blood Microbiota Implicated in COVID-19 Prognosis. Cells 2021, 10, 1452. [Google Scholar] [CrossRef]
  20. Petruk, G.; Puthia, M.; Petrlova, J.; Samsudin, F.; Strömdahl, A.C.; Cerps, S.; Uller, L.; Kjellström, S.; Bond, P.J.; Schmidtchen, A.A. SARS-CoV-2 spike protein binds to bacterial lipopolysaccharide and boosts proinflammatory activity. J. Mol. Cell Biol. 2020, 12, 916–932. [Google Scholar] [CrossRef]
  21. Kaushik, D.; Bhandari, R.; Kuhad, A. TLR4 as a therapeutic target for respiratory and neurological complications of SARS-CoV-2. Expert Opin. Ther. Targets 2021, 25, 491–508. [Google Scholar] [CrossRef] [PubMed]
  22. Wenzhong, L.; Hualan, L. COVID-19: The CaMKII-like system of S protein drives membrane fusion and induces syncytial multinucleated giant cells. Immunol. Res. 2021, 69, 496–519. [Google Scholar] [CrossRef] [PubMed]
  23. Ghosh, A.; Giese, K.P. Calcium/calmodulin-dependent kinase II and Alzheimer’s disease. Mol. Brain 2015, 8, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. De Dios, L.; Collazo, C.; Inostroza-Nieves, Y. Renin-angiotensin-system increases phosphorylated tau and Reactive Oxygen Species in human cortical neuron cell line. Biochem. Biophys. Rep. 2022, 32, 101355. [Google Scholar] [CrossRef] [PubMed]
  25. Giordano-Santin, R.; Kaulich, E.; Galbraith, K.M.; Hilliard, M.A. Fusogen-mediated neuron−neuron fusion disrupts neural circuit connectivity and alters animal behavior. Proc. Natl. Acad. Sci. USA 2020, 117, 23054–23065. [Google Scholar] [CrossRef] [PubMed]
  26. Faizan, M.I.; Chaudhuri, R.; Sagar, S.; Albogami, S.; Chaudhary, N.; Azmi, I.; Akhtar, A.; Ali, S.M.; Kumar, R.; Iqbal, J.; et al. NSP4 and ORF9b of SARS-CoV-2 Induce Pro-Inflammatory Mitochondrial DNA Release in Inner MembraneDerived Vesicles. Cells 2022, 11, 2969. [Google Scholar] [CrossRef]
  27. Pliss, A.; Kuzmin, A.N.; Prasad, P.N.; Mahajan, S.D. Mitochondrial Dysfunction: A Prelude to Neuropathogenesis of SARS-CoV-2. ACS Chem. Neurosci. 2022, 13, 308–312. [Google Scholar] [CrossRef]
  28. Duan, C.; Ma, R.; Zeng, X.; Chen, B.; Hou, D.; Liu, R.; Li, X.; Liu, L.; Li, T.; Huang, H. SARS-CoV-2 Achieves Immune Escape by Destroying Mitochondrial Quality: Comprehensive Analysis of the Cellular Landscapes of Lung and Blood Specimens from Patients With COVID-19. Front. Immunol. 2022, 13, 946731. [Google Scholar] [CrossRef]
  29. Giordano-Santini, R.; Linton, C.; Hilliard, M.A. Cell-cell fusion in the nervous system: Alternative mechanisms of development, injury, and repair. Semin. Cell Dev. Biol. 2016, 60, 146–154. [Google Scholar] [CrossRef] [Green Version]
  30. Barthélemy, N.R.; Horie, K.; Sato, C.; Bateman, R.J. Blood plasma phosphorylated-tau isoforms track CNS change in Alzheimer’s disease. J. Exp. Med. 2020, 217, e20200861, PMCID:PMC7596823. [Google Scholar] [CrossRef] [PubMed]
  31. Sotnikov, O.S. Binucleated and Multinucleated Neurons are Formed by Fusion. Bull. Exp. Biol. Med. 2021, 171, 508–512. [Google Scholar] [CrossRef]
  32. Martin, N.; Bernard, D. Calcium signaling and cellular senescence. Cell Calcium 2018, 70, 16–23. [Google Scholar] [CrossRef]
  33. Whitlock, J.M.; Chernomordik, L.V. Flagging fusion: Phosphatidylserine signaling in cell-cell fusion. J. Biol. Chem. 2021, 296, 100411. [Google Scholar] [CrossRef] [PubMed]
  34. Arora, A.; Kolberg, J.E.; Badarinarayan, S.S.; Munot, D.; Müller, M.; Sauter, D.; Bansal, V. SARS-CoV-2 infection activates endogenous retroviruses of the LTR69 subfamily. bioRxiv 2023. [Google Scholar] [CrossRef]
  35. Grandi, N.; Erbì, M.C.; Scognamiglio, S.; Tramontano, E. Human Endogenous Retrovirus (HERV) Transcriptome Is Dynamically Modulated during SARS-CoV-2 Infection and Allows Discrimination of COVID-19 Clinical Stages. Microbiol. Spectr. 2023, 11, e0251622. [Google Scholar] [CrossRef] [PubMed]
  36. Gonzalez-Cao, M.; Iduma, P.; Karachaliou, N.; Santarpia, M.; Blanco, J.; Rosell, R. Human endogenous retroviruses and cancer. Cancer Biol. Med. 2016, 13, 483–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Shukla, R.; Santoro, J.; Bender, F.C.; Laterza, O.F. Quantitative determination of human interleukin 22 (IL-22) in serum using Singulex-Erenna® technology. J. Immunol. Methods 2013, 390, 30–34. [Google Scholar] [CrossRef]
  38. Eichenberger, E.M.; Degner, N.; Scott, E.R.; Ruffin, F.; Franzone, J.; Sharma-Kuinkel, B.; Shah, P.; Hong, D.; Dalai, S.C.; Blair, L.; et al. Microbial Cell-Free DNA Identifies the Causative Pathogen in Infective Endocarditis and Remains Detectable Longer Than Conventional Blood Culture in Patients with Prior Antibiotic Therapy. Clin. Infect. Dis. 2022, 76, e1492–e1500. [Google Scholar] [CrossRef]
  39. Thijssen, E.H.; La Joie, R.; Strom, A.; Fonseca, C.; Iaccarino, L.; Wolf, A.; Spina, S.; Allen, I.E.; Cobigo, Y.; Heuer, H.; et al. Advancing Research and Treatment for Frontotemporal Lobar Degeneration investigators. Plasma phosphorylated tau 217 and phosphorylated tau 181 as biomarkers in Alzheimer’s disease and frontotemporal lobar degeneration: A retrospective diagnostic performance study. Lancet Neurol. 2021, 20, 739–752. [Google Scholar] [CrossRef]
  40. Bayoumy, S.; Verberk, I.M.W.; Dulk, B.D.; Hussainali, Z.; Zwan, M.; van der Flier, W.M.; Ashton, N.J.; Zetterberg, H.; Blennow, K.; Vanbrabant, J.; et al. Clinical and analytical comparison of six Simoa assays for plasma P-tau isoforms P-tau181, P-tau217, and P-tau231. Alzheimer’s Res. Ther. 2021, 13, 198. [Google Scholar] [CrossRef]
  41. Eberth, J. About the finer construction of the intestinal mucosa. In Wurzb Naturwiss Zeitschr; Stahel’schen Buch- und Kunsthandlung: Würzburg, Germany, 1864; Volume 5. [Google Scholar]
  42. Patterson, B.K.; Francisco, E.B.; Yogendra, R.; Long, E.; Pise, A.; Rodrigues, H.; Hall, E.; Herrera, M.; Parikh, P.; Guevara-Coto, J.; et al. Persistence of SARS-CoV-2 S1 Protein in CD16+ Monocytes in Post-Acute Sequelae of COVID-19 (PASC) up to 15 Months Post-Infection. Front. Immunol. 2022, 12, 746021. [Google Scholar] [CrossRef]
  43. Ahamed, J.; Laurence, J. Long COVID endotheliopathy: Hypothesized mechanisms and potential therapeutic approaches. J. Clin. Investig. 2022, 132, e161167. [Google Scholar] [CrossRef]
  44. Monje, M.; Iwasaki, A. The neurobiology of long COVID. Neuron 2022, 110, 3484–3496. [Google Scholar] [CrossRef]
  45. Zhu, J.; Wilding, J.P.H.; Hu, J. Adipocytes in obesity: A perfect reservoir for SARS-CoV-2? Med. Hypotheses 2023, 171, 111020. [Google Scholar] [CrossRef] [PubMed]
  46. Neurath, M.F.; Überla, K.; Ng, S.C. Gut as viral reservoir: Lessons from gut viromes, HIV and COVID-19. Gut 2021, 70, 1605–1608. [Google Scholar] [CrossRef] [PubMed]
  47. Ruscitti, P.; Ursini, F.; Shoenfeld, Y. Ferritin and myalgic encephalomyelitis/chronic fatigue syndrome in post COVID-19, an unexpected facet of the hyperferritinemic syndrome? J. Psychosom. Res. 2023, 169, 111231. [Google Scholar] [CrossRef] [PubMed]
  48. Sun, B.; Tang, N.; Peluso, M.J.; Iyer, N.S.; Torres, L.; Donatelli, J.L.; Munter, S.E.; Nixon, C.C.; Rutishauser, R.L.; Rodriguez-Barraquer, I.; et al. Characterization and Biomarker Analyses of Post-COVID-19 Complications and Neurological Manifestations. Cells 2021, 10, 386. [Google Scholar] [CrossRef]
  49. Wallet, C.; De Rovere, M.; Van Assche, J.; Daouad, F.; De Wit, S.; Gautier, V.; Mallon, P.W.G.; Marcello, A.; Van Lint, C.; Rohr, O.; et al. Microglial Cells: The Main HIV-1 Reservoir in the Brain. Front. Cell. Infect. Microbiol. 2019, 9, 362. [Google Scholar] [CrossRef] [Green Version]
  50. Horkowitz, A.P.; Schwartz, A.V.; Alvarez, C.A.; Herrera, E.B.; Thoman, M.L.; Chatfield, D.A.; Osborn, K.G.; Feuer, R.; George, U.Z.; Phillips, J.A. Acetylcholine Regulates Pulmonary Pathology During Viral Infection and Recovery. ImmunoTargets Ther. 2020, 9, 333–350. [Google Scholar] [CrossRef]
  51. Angulo, O.; Restrepo, H.F.; Higuera, J.G.L.; Garcia, M.L.; Mendive, F.; Politi, M. Effectiveness in humans of an antiviral drug based on cholinergic agonists with specific design and route of administration against COVID-19 symptoms in a group of infected patients vs. Control group. Qeios 2021. preprint. [Google Scholar] [CrossRef]
  52. Jafarzadeh, A.; Nemati, M.; Jafarzadeh, S. Contribution of STAT3 to the pathogenesis of COVID-19. Microb. Pathog. 2021, 154, 104836. [Google Scholar] [CrossRef]
  53. Matsuyama, T.; Kubli, S.P.; Yoshinaga, S.K.; Pfeffer, K.; Mak, T.W. An aberrant STAT pathway is central to COVID-19. Cell Death Differ. 2020, 27, 3209–3225. [Google Scholar] [CrossRef] [PubMed]
  54. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; McCarthy, P.; Lange, F.; Andersson, J.L.R.; Griffanti, L.; Duff, E.; et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022, 604, 697–707. [Google Scholar] [CrossRef] [PubMed]
  55. Rothstein, T.L. Cortical Grey matter volume depletion links to neurological sequelae in post COVID-19 “long haulers”. BMC Neurol. 2023, 23, 22. [Google Scholar] [CrossRef]
  56. Mazza, M.G.; Palladini, M.; De Lorenzo, R.; Magnaghi, C.; Poletti, S.; Furlan, R.; Ciceri, F.; COVID-19 BioB Outpatient Clinic Study Group; Rovere-Querini, P.; Benedetti, F. Persistent psychopathology and neurocognitive impairment in COVID-19 survivors: Effect of inflammatory biomarkers at three-month follow-up. Brain Behav. Immun. 2021, 94, 138–147. [Google Scholar] [CrossRef]
  57. Andrews, M.G.; Mukhtar, T.; Eze, U.C.; Simoneau, C.R.; Ross, J.; Parikshak, N.; Wang, S.; Zhou, L.; Koontz, M.; Velmeshev, D.; et al. Tropism of SARS-CoV-2 for human cortical astrocytes. Proc. Natl. Acad. Sci. USA 2022, 119, e2122236119. [Google Scholar] [CrossRef] [PubMed]
  58. Avolio, E.; Carrabba, M.; Milligan, R.; Williamson, M.K.; Beltrami, A.P.; Gupta, K.; Elvers, K.T.; Gamez, M.; Foster, R.R.; Gillespie, K.; et al. The SARS-CoV-2 Spike protein disrupts human cardiac pericytes function through CD147 receptor-mediated signalling: A potential non-infective mechanism of COVID-19 microvascular disease. Clin. Sci. 2021, 135, 2667–2689. [Google Scholar] [CrossRef]
  59. Parpura, V.; Verkhratsky, A. Astrocytes revisited: Concise historic outlook on glutamate homeostasis and signaling. Croat. Med J. 2012, 53, 518–528. [Google Scholar] [CrossRef] [Green Version]
  60. Valdebenito, S.; Castellano, P.; Ajasin, D.; Eugenin, E.A. Astrocytes are HIV reservoirs in the brain: A cell type with poor HIV infectivity and replication but efficient cell-to-cell viral transfer. J. Neurochem. 2021, 158, 429–443. [Google Scholar] [CrossRef]
  61. Huang, L.; Nakamura, Y.; Lo, E.H.; Hayakawa, K. Astrocyte Signaling in the Neurovascular Unit After Central Nervous System Injury. Int. J. Mol. Sci. 2019, 20, 282. [Google Scholar] [CrossRef] [Green Version]
  62. Tang, Y.; Chaillon, A.; Gianella, S.; Wong, L.M.; Li, D.; Simermeyer, T.L.; Porrachia, M.; Ignacio, C.; Woodworth, B.; Zhong, D.; et al. Brain microglia serve as a persistent HIV reservoir despite durable antiretroviral therapy. J. Clin. Investig. 2023, 133, e167417. [Google Scholar] [CrossRef]
  63. Lagoumintzis, G.; Chasapis, C.T.; Alexandris, N.; Kouretas, D.; Tzartos, S.; Eliopoulos, E.; Farsalinos, K.; Poulas, K. Nicotinic cholinergic system and COVID-19: In silico identification of interactions between α7 nicotinic acetylcholine receptor and the cryptic epitopes of SARS-Co-V and SARS-CoV-2 Spike glycoproteins. Food Chem. Toxicol. 2021, 149, 112009. [Google Scholar] [CrossRef]
  64. Terry, A.V., Jr.; Callahan, P.M. α7 nicotinic acetylcholine receptors as therapeutic targets in schizophrenia: Update on animal and clinical studies and strategies for the future. Neuropharmacology 2020, 170, 108053. [Google Scholar] [CrossRef]
  65. Zhao, X.; Wilson, K.; Uteshev, V.; He, J.J. Activation of α7 nicotinic acetylcholine receptor ameliorates HIV-associated neurology and neuropathology. Brain 2021, 144, 3355–3370. [Google Scholar] [CrossRef] [PubMed]
  66. Zeng, C.; Evans, J.P.; King, T.; Zheng, Y.M.; Oltz, E.M.; Whelan, S.P.J.; Saif, L.J.; Peeples, M.E.; Liu, S.L. SARS-CoV-2 spreads through cell-to-cell transmission. Proc. Natl. Acad. Sci. USA 2021, 119, e2111400119. [Google Scholar] [CrossRef]
  67. Masaldan, S.; Clatworthy, S.A.; Gamell, C.; Meggyesy, P.M.; Rigopoulos, A.-T.; Haupt, S.; Haupt, Y.; Denoyer, D.; Adlard, P.A.; Bush, A.I.; et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 2018, 14, 100–115. [Google Scholar] [CrossRef] [PubMed]
  68. Wiel, C.; Lallet-Daher, H.; Gitenay, D.; Gras, B.; Le Calvé, B.; Augert, A.; Ferrand, M.; Prevarskaya, N.; Simonnet, H.; Vindrieux, D.; et al. Endoplasmic reticulum calcium release through ITPR2 channels leads to mitochondrial calcium accumulation and senescence. Nat. Commun. 2014, 5, 3792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Birge, R.B.; Boeltz, S.; Kumar, S.; Carlson, J.; Wanderley, J.; Calianese, D.; Barcinski, M.; Brekken, R.A.; Huang, X.; Hutchins, J.T.; et al. Phosphatidylserine is a global immunosuppressive signal in efferocytosis, infectious disease, and cancer. Cell Death Differ. 2016, 23, 962–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Hassanzadeh, K.; Perez Pena, H.; Dragotto, J.; Buccarello, L.; Iorio, F.; Pieraccini, S.; Sancini, G.; Feligioni, M. Considerations around the SARS-CoV-2 Spike Protein with Particular Attention to COVID-19 Brain Infection and Neurological Symp-toms. ACS Chem. Neurosci. 2020, 11, 2361–2369. [Google Scholar] [CrossRef]
  71. Bourque, G.; Burns, K.H.; Gehring, M.; Gorbunova, V.; Seluanov, A.; Hammell, M.; Imbeault, M.; Izsvák, Z.; Levin, H.L.; Macfarlan, T.S.; et al. Ten things you should know about transposable elements. Genome Biol. 2018, 19, 199. [Google Scholar] [CrossRef]
  72. Nelson, P.N.; Hooley, P.; Roden, D.; Davari Ejtehadi, H.; Rylance, P.; Warren, P.; Martin, J.; Murray, P.G.; Molecular Immunology Research Group. Human endogenous retroviruses: Transposable elements with potential? Clin. Exp. Immunol. 2004, 138, 1–9. [Google Scholar] [CrossRef]
  73. Wang, J.; Zuo, X.; Dai, Z.; Xia, M.; Zhao, Z.; Zhao, X.; Jia, J.; Han, Y.; He, Y. Disrupted functional brain connectome in individuals at risk for Alzheimer’s disease. Biol Psychiatry. 2013, 73, 472–481. [Google Scholar] [CrossRef] [PubMed]
  74. Sfera, A.; Bullock, K.; Price, A.; Inderias, L.; Osorio, C. Ferrosenescence: The iron age of neurodegeneration? Mech. Ageing Dev. 2018, 174, 63–75. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, J.; Chen, X. p53 tumor suppressor and iron homeostasis. FEBS J. 2019, 286, 620–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Tian, T.; Wu, J.; Chen, T.; Li, J.; Yan, S.; Zhou, Y.; Peng, X.; Li, Y.; Zheng, N.; Cai, A.; et al. Long-term follow-up of dynamic brain changes in patients recovered from COVID-19 without neurological manifestations. J. Clin. Investig. Insight 2022, 7, e155827. [Google Scholar] [CrossRef]
  77. Sonnweber, T.; Boehm, A.; Sahanic, S.; Pizzini, A.; Aichner, M.; Sonnweber, B.; Kurz, K.; Koppelstätter, S.; Haschka, D.; Petzer, V.; et al. Persisting alterations of iron homeostasis in COVID-19 are associated with non-resolving lung pathologies and poor patients’ performance: A prospective observational cohort study. Respir. Res. 2020, 21, 276. [Google Scholar] [CrossRef]
  78. Courrol, L.C.; de Oliveira Silva, F.R.; Masilamani, V. SARS-CoV-2, hemoglobin and protoporphyrin IX: Interactions and perspectives. Photodiagn. Photodyn. Ther. 2021, 34, 102324. [Google Scholar] [CrossRef]
  79. Singh, N.; Bharara Singh, A. S2 subunit of SARS-nCoV-2 interacts with tumor suppressor protein p53 and BRCA: An in silico study. Transl. Oncol. 2020, 13, 100814. [Google Scholar] [CrossRef]
  80. Martínez-Mármol, R.; Giordano-Santini, R.; Kaulich, E.; Cho, A.-N.; Przybyla, M.; Riyadh, A.; Robinson, E.; Chew, K.Y.; Amor, R.; Meunier, F.A.; et al. SARS-CoV-2 infection and viral fusogens cause neuronal and glial fusion that compromises neuronal activity. Sci. Adv. 2023, 9, eadg2248. [Google Scholar] [CrossRef]
  81. Tang, J.; Frascaroli, G.; Zhou, X.; Knickmann, J.; Brune, W. Cell Fusion and Syncytium Formation in Betaherpesvirus Infection. Viruses 2021, 13, 1973. [Google Scholar] [CrossRef]
  82. Liu, S.; Hossinger, A.; Heumüller, S.-E.; Hornberger, A.; Buravlova, O.; Konstantoulea, K.; Müller, S.A.; Paulsen, L.; Rousseau, F.; Schymkowitz, J.; et al. Highly efficient intercellular spreading of protein misfolding mediated by viral ligand-receptor interactions. Nat. Commun. 2021, 12, 5739. [Google Scholar]
  83. Chakraborty, C.; Bhattacharya, M.; Sharma, A.R. Present variants of concern and variants of interest of severe acute respiratory syndrome coronavirus 2: Their significant mutations in S-glycoprotein, infectivity, re-infectivity, immune escape and vaccines activity. Rev. Med Virol. 2021, 32, e2270. [Google Scholar] [CrossRef]
  84. Pavelka, M.; Roth, J. Myofibrils, Intercalated Disk. In Functional Ultrastructure; Springer: Vienna, Austria, 2010. [Google Scholar] [CrossRef]
  85. Guller, S.; Ma, Y.Y.; Fu, H.H.; Krikun, G.; Abrahams, V.M.; Mor, G. The placental syncytium and the pathophysiology of preeclampsia and intrauterine growth restriction: A novel assay to assess syncytial protein expression. Ann. N. Y. Acad. Sci. 2008, 1127, 129–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Campa, V.M.; Gutiérrez-Lanza, R.; Cerignoli, F.; Díaz-Trelles, R.; Nelson, B.; Tsuji, T.; Barcova, M.; Jiang, W.; Mercola, M. Notch activates cell cycle reentry and progression in quiescent cardiomyocytes. J. Cell Biol. 2008, 183, 129–141. [Google Scholar] [CrossRef]
  87. Silva, C.C.; Bichara, C.N.C.; Carneiro, F.R.O.; Palacios, V.R.D.C.M.; Berg, A.V.S.V.D.; Quaresma, J.A.S.; Magno Falcão, L.F. Muscle dysfunction in the long coronavirus disease 2019 syndrome: Pathogenesis and clinical approach. Rev. Med Virol. 2022, 32, e2355. [Google Scholar] [CrossRef]
  88. Nagy, J.I.; Rash, J.E. Connexins and gap junctions of astrocytes and oligodendrocytes in the CNS. Brain Res. Rev. 2000, 32, 29–44. [Google Scholar] [CrossRef] [PubMed]
  89. Faggioli, F.; Wang, T.; Vijg, J.; Montagna, C. Chromosome-specific accumulation of aneuploidy in the aging mouse brain. Hum. Mol. Genet. 2012, 21, 5246–5253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Caneus, J.; Granic, A.; Rademakers, R.; Dickson, D.W.; Coughlan, C.M.; Chial, H.J.; Potter, H. Mitotic defects lead to neuronal aneuploidy and apoptosis in frontotemporal lobar degeneration caused by MAPT mutations. Mol. Biol. Cell 2018, 29, 575–586. [Google Scholar] [CrossRef]
  91. Kiyoshi, C.M.; Zhou, M. Astrocyte syncytium: A functional reticular system in the brain. Neural Regen. Res. 2019, 14, 595–596. [Google Scholar] [CrossRef]
  92. Sotelo, C. Camillo Golgi and Santiago Ramon y Cajal: The anatomical organization of the cortex of the cerebellum. Can the neuron doctrine still support our actual knowledge on the cerebellar structural arrangement? Brain Res. Rev. 2011, 66, 16–34. [Google Scholar] [CrossRef] [Green Version]
  93. Alvarez-Dolado, M.; Pardal, R.; Garcia-Verdugo, J.M.; Fike, J.R.; Lee, H.O.; Pfeffer, K.; Lois, C.; Morrison, S.J.; Alvarez-Buylla, A. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nat. Cell Biol. 2003, 425, 968–973. [Google Scholar] [CrossRef] [Green Version]
  94. Ishunina, T.A.; Unmehopa, U.A.; Dolzhikov, A.A.; Swaab, D.F. Multinucleated Arginine-Vasopressin Neurons in the Human Supraoptic Nucleus: A Hallmark of Pulmonary Pathology. Neuroendocrinology 2000, 72, 318–326. [Google Scholar] [CrossRef] [PubMed]
  95. Kemp, K.; Wilkins, A.; Scolding, N. Cell fusion in the brain: Two cells forward, one cell back. Acta Neuropathol. 2014, 128, 629–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Hornik, T.C.; Neniskyte, U.; Brown, G.C. Inflammation Induces Multinucleation of Microglia via PKC Inhibition of Cytokinesis, Generating Highly Phagocytic Multinucleated Giant Cells. J. Neurochem. 2013, 128, 650–661. [Google Scholar] [CrossRef] [PubMed]
  97. Iourov, I.Y.; Heng, H.H. Editorial: Somatic genomic mosaicism & human disease. Front. Genet. 2022, 13, 1045559. [Google Scholar] [CrossRef] [PubMed]
  98. Griffiths, D.J. Endogenous retroviruses in the human genome sequence. Genome Biol. 2001, 2, REVIEWS1017. [Google Scholar] [CrossRef]
  99. Chang, N.T.; Yang, W.K.; Huang, H.C.; Yeh, K.W.; Wu, C.W. The transcriptional activity of HERV-I LTR is negatively regulated by its cis-elements and wild type p53 tumor suppressor protein. J. Biomed. Sci. 2006, 14, 211–222. [Google Scholar] [CrossRef]
  100. Brooks, C.L.; Gu, W. p53 ubiquitination: Mdm2 and beyond. Mol. Cell 2006, 21, 307–315. [Google Scholar] [CrossRef] [Green Version]
  101. Dörnen, J.; Sieler, M.; Weiler, J.; Keil, S.; Dittmar, T. Cell Fusion-Mediated Tissue Regeneration as an Inducer of Polyploidy and Aneuploidy. Int. J. Mol. Sci. 2020, 21, 1811. [Google Scholar] [CrossRef] [Green Version]
  102. Melano, I.; Kuo, L.-L.; Lo, Y.-C.; Sung, P.-W.; Tien, N.; Su, W.-C. Effects of Basic Amino Acids and Their Derivatives on SARS-CoV-2 and Influenza-A Virus Infection. Viruses 2021, 13, 1301. [Google Scholar] [CrossRef]
  103. Allolio, C.; Magarkar, A.; Jurkiewicz, P.; Baxová, K.; Javanainen, M.; Mason, P.E.; Šachl, R.; Cebecauer, M.; Hof, M.; Horinek, D.; et al. Arginine-Rich Cell-Penetrating Peptides Induce Membrane Multilamellarity and Subsequently Enter via Formation of a Fusion Pore. Proc. Natl. Acad. Sci. USA 2018, 115, 11923–11928. [Google Scholar] [CrossRef] [Green Version]
  104. Nyenhuis, S.B.; Karandikar, N.; Kiessling, V.; Kreutzberger, A.J.B.; Thapa, A.; Liang, B.; Tamm, L.K.; Cafiso, D.S. Conserved Arginine Residues in Synaptotagmin 1 Regulate Fusion Pore Expansion Through Membrane Contact. Nat. Commun. 2021, 12, 761. [Google Scholar] [CrossRef] [PubMed]
  105. Al-Motawa, M.S.; Abbas, H.; Wijten, P.; de la Fuente, A.; Xue, M.; Rabbani, N.; Thornalley, P.J. Vulnerabilities of the SARS-CoV-2 Virus to Proteotoxicity-Opportunity for Repurposed Chemotherapy of COVID-19 Infection. Front. Pharmacol. 2020, 11, 585408. [Google Scholar] [CrossRef] [PubMed]
  106. Knerr, I.; Beinder, E.; Rascher, W. Syncytin, a novel human endogenous retroviral gene in human placenta: Evidence for its dysregulation in preeclampsia and HELLP syndrome. Am. J. Obstet. Gynecol. 2002, 186, 210–213. [Google Scholar] [CrossRef] [PubMed]
  107. Vance, T.D.R.; Lee, J.E. Virus and eukaryote fusogen superfamilies. Curr. Biol. 2020, 30, R750–R754. [Google Scholar] [CrossRef]
  108. Tug, E.; Yirmibes Karaoguz, M.; Nas, T. Expression of the syncytin-1 and syncytin-2 genes in the trophoblastic tissue of the early pregnancy losses with normal and abnormal karyotypes. Gene 2020, 741, 144533. [Google Scholar] [CrossRef]
  109. Pastuzyn, E.D.; Day, C.E.; Kearns, R.B.; Kyrke-Smith, M.; Taibi, A.V.; McCormick, J.; Yoder, N.; Belnap, D.M.; Erlendsson, S.; Morado, D.R.; et al. The Neuronal Gene Arc Encodes a Repurposed Retrotransposon Gag Protein that Mediates Intercellular RNA Transfer. Cell 2018, 172, 275–288.e18, Erratum in Cells 2018, 173, 275. [Google Scholar] [CrossRef] [Green Version]
  110. Liu, M.; Jia, L.; Li, H.; Liu, Y.; Han, J.; Wang, X.; Li, T.; Li, J.; Zhang, B.; Zhai, X.; et al. p53 Binding Sites in Long Terminal Repeat 5Hs (LTR5Hs) of Human Endogenous Retrovirus K Family (HML-2 Subgroup) Play Important Roles in the Regulation of LTR5Hs Transcriptional Activity. Microbiol. Spectr. 2022, 10, e0048522. [Google Scholar] [CrossRef]
  111. Ma-Lauer, Y.; Carbajo-Lozoya, J.; Hein, M.Y.; Müller, M.A.; Deng, W.; Lei, J.; Meyer, B.; Kusov, Y.; von Brunn, B.; Bairad, D.R.; et al. p53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PLpro via E3 ubiquitin ligase RCHY1. Proc. Natl. Acad. Sci. USA 2016, 113, E5192–E5201. [Google Scholar] [CrossRef]
  112. Wang, X.; Huang, J.; Zhu, F. Human Endogenous Retroviral Envelope Protein Syncytin-1 and Inflammatory Abnormalities in Neuropsychological Diseases. Front. Psychiatry 2018, 9, 422. [Google Scholar] [CrossRef]
  113. Nekrutenko, A.; Li, W.H. Transposable elements are found in a large number of human protein-coding genes. Trends Genet. 2001, 17, 619–621. [Google Scholar] [CrossRef]
  114. Gómez-Carballa, A.; Martinón-Torres, F.; Salas, A. Is SARS-CoV-2 an oncogenic virus? J. Infect. 2022, 85, 573–607. [Google Scholar] [CrossRef] [PubMed]
  115. Costanzo, M.; De Giglio, M.A.R.; Roviello, G.N. Deciphering the Relationship between SARS-CoV-2 and Cancer. Int. J. Mol. Sci. 2023, 24, 7803. [Google Scholar] [CrossRef] [PubMed]
  116. Saini, G.; Aneja, R. Cancer as a prospective sequela of long COVID-19. Bioessays 2021, 43, e2000331. [Google Scholar] [CrossRef] [PubMed]
  117. Polozov, S.; Cruz-Garcia, L.; O’Brien, G.; Goriacha, V.; Nasser, F.; Jeggo, P.; Candéias, S.; Badie, C. Deficient Radiation Transcription Response in COVID-19 Patients. Adv. Radiat. Oncol. 2023, 8, 101215. [Google Scholar] [CrossRef] [PubMed]
  118. Kim, C.J.; Nazli, A.; Rojas, O.L.; Chege, D.; Alidina, Z.; Huibner, S.; Mujib, S.; Benko, E.; Kovacs, C.; Shin, L.Y.Y.; et al. A role for mucosal IL-22 production and Th22 cells in HIV-associated mucosal immunopathogenesis. Mucosal Immunol. 2012, 5, 670–680. [Google Scholar] [CrossRef] [PubMed]
  119. Shi, M.; Zong, X.; Hur, J.; Birmann, B.M.; Martinez-Maza, O.; Epeldegui, M.; Chan, A.T.; Giovannucci, E.L.; Cao, Y. Circulating markers of microbial translocation and host response to bacteria with risk of colorectal cancer: A prospective, nested case-control study in men. Ebiomedicine 2023, 91, 104566. [Google Scholar] [CrossRef]
  120. Sun, Q.; Chen, W. Cell-in-cell: An Emerging Player in COVID-19 and Immune Disorders. Natl. Sci. Open 2022, 1, 1. [Google Scholar]
  121. Bauer, M.F.; Hader, M.; Hecht, M.; Büttner-Herold, M.; Fietkau, R.; Distel, L.V.R. Cell-in-cell phenomenon: Leukocyte engulfment by non-tumorigenic cells and cancer cell lines. BMC Cell Biol. 2021, 22, 39. [Google Scholar] [CrossRef]
  122. Dudakov, J.A.; Hanash, A.M.; van den Brink, M.R. Interleukin-22: Immunobiology and pathology. Annu. Rev. Immunol. 2015, 33, 747–785. [Google Scholar] [CrossRef] [Green Version]
  123. Keir, M.; Yi, Y.; Lu, T.; Ghilardi, N. The role of IL-22 in intestinal health and disease. J. Exp. Med. 2020, 217, e20192195. [Google Scholar] [CrossRef]
  124. Arshad, T.; Mansur, F.; Palek, R.; Manzoor, S.; Liska, V. A Double Edged Sword Role of Interleukin-22 in Wound Healing and Tissue Regeneration. Front. Immunol. 2020, 11, 2148. [Google Scholar] [CrossRef] [PubMed]
  125. Ezeonwumelu, I.J.; Garcia-Vidal, E.; Ballana, E. JAK-STAT Pathway: A Novel Target to Tackle Viral Infections. Viruses 2021, 13, 2379. [Google Scholar] [CrossRef] [PubMed]
  126. Subbanna, M.; Shivakumar, V.; Talukdar, P.M.; Narayanaswamy, J.C.; Venugopal, D.; Berk, M.; Varambally, S.; Venkatasubramanian, G.; Debnath, M. Role of IL-6/RORC/IL-22 axis in driving Th17 pathway mediated immunopathogenesis of schizophrenia. Cytokine 2018, 111, 112–118. [Google Scholar] [CrossRef] [PubMed]
  127. Barthelemy, A.; Sencio, V.; Soulard, D.; Deruyter, L.; Faveeuw, C.; Le Goffic, R.; Trottein, F. Interleukin-22 Immunotherapy during Severe Influenza Enhances Lung Tissue Integrity and Reduces Secondary Bacterial Systemic Invasion. Infect. Immun. 2018, 86, e00706-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Albayrak, N.; Orte Cano, C.; Karimi, S.; Dogahe, D.; Van Praet, A.; Godefroid, A.; Del Marmol, V.; Grimaldi, D.; Bondue, B.; Van Vooren, J.P.; et al. Distinct Expression Patterns of Interleukin-22 Receptor 1 on Blood Hematopoietic Cells in SARS-CoV-2 Infection. Front. Immunol. 2022, 13, 769839. [Google Scholar] [CrossRef] [PubMed]
  129. Li, L.J.; Gong, C.; Zhao, M.H.; Feng, B.S. Role of interleukin-22 in inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 18177–18188. [Google Scholar] [CrossRef]
  130. Rajapaksha, P.; Elbourne, A.; Gangadoo, S.; Brown, R.; Cozzolino, D.; Chapman, J. A Review of Methods for the Detection of Pathogenic Microorganisms. Anayst 2018, 144, 396–411. [Google Scholar] [CrossRef]
  131. Park, S.Y.; Chang, E.J.; Ledeboer, N.; Messacar, K.; Lindner, M.S.; Venkatasubrahmanyam, S.; Wilber, J.C.; Vaughn, M.L.; Perkins, B.A.; Nolte, F.S. Plasma Microbial Cell-free DNA Sequencing from Over 15,000 Patients Identified a Broad Spectrum of Pathogens. medRxiv 2023. [Google Scholar] [CrossRef]
  132. Chiocchetti, R.; Hitrec, T.; Giancola, F.; Sadeghinezhad, J.; Squarcio, F.; Galiazzo, G.; Piscitiello, E.; De Silva, M.; Cerri, M.; Amici, R.; et al. Phosphorylated Tau protein in the myenteric plexus of the ileum and colon of normothermic rats and during synthetic torpor. Cell Tissue Res. 2021, 384, 287–299. [Google Scholar] [CrossRef]
  133. Prigent, A.; Chapelet, G.; Lataillade, A.D.G.D.; Oullier, T.; Durieu, E.; Bourreille, A.; Duchalais, E.; Hardonnière, K.; Neunlist, M.; Noble, W.; et al. Tau accumulates in Crohn’s disease gut. FASEB J. 2020, 34, 9285–9296. [Google Scholar] [CrossRef]
  134. Ferrara, F.; Zovi, A.; Masi, M.; Langella, R.; Trama, U.; Boccellino, M.; Vitiello, A. Long COVID could become a widespread post-pandemic disease? A debate on the organs most affected. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 1583–1589. [Google Scholar] [CrossRef] [PubMed]
  135. Song, M.; Graubard, B.I.; Rabkin, C.S.; Engels, E.A. Neutrophil-to-lymphocyte ratio and mortality in the United States general population. Sci. Rep. 2021, 11, 464. [Google Scholar] [CrossRef] [PubMed]
  136. Ryan, F.J.; Hope, C.M.; Masavuli, M.G.; Lynn, M.A.; Mekonnen, Z.A.; Yeow, A.E.L.; Garcia-Valtanen, P.; Al-Delfi, Z.; Gummow, J.; Ferguson, C.; et al. Long-term perturbation of the peripheral immune system months after SARS-CoV-2 infection. BMC Med. 2022, 20, 26. [Google Scholar] [CrossRef]
  137. Li, Q.; Wang, Y.; Sun, Q.; Knopf, J.; Herrmann, M.; Lin, L.; Jiang, J.; Shao, C.; Li, P.; He, X.; et al. Immune response in COVID-19: What is next. Cell Death Differ. 2022, 29, 1107–1122. [Google Scholar] [CrossRef] [PubMed]
  138. Rex, D.A.B.; Prasad, T.S.K.; Kandasamy, R.K. Revisiting Regulated Cell Death Responses in Viral Infections. Int. J. Mol. Sci. 2022, 23, 7023. [Google Scholar] [CrossRef] [PubMed]
  139. Alosaimi, M.F.; Shendi, H.; Beano, A.; Stafstrom, K.; El Hawary, R.; Meshaal, S.; Galal, N.; Pai, S.Y.; El-Marsafy, A.; Geha, R.S.; et al. T-cell mitochondrial dysfunction and lymphopenia in DOCK2-deficient patients. J. Allergy Clin. Immunol. 2019, 144, 306–309.e2. [Google Scholar] [CrossRef] [Green Version]
  140. Zhang, Y.; Zhang, J.; Duan, L. The role of microbiota-mitochondria crosstalk in pathogenesis and therapy of intestinal diseases. Pharmacol. Res. 2022, 186, 106530. [Google Scholar] [CrossRef]
  141. Kopańska, M.; Batoryna, M.; Bartman, P.; Szczygielski, J.; Banaś-Ząbczyk, A. Disorders of the Cholinergic System in COVID-19 Era-A Review of the Latest Research. Int. J. Mol. Sci. 2022, 23, 672. [Google Scholar] [CrossRef]
  142. Hübers, A.; Lascano, A.M.; Lalive, P.H. Management of patients with generalised myasthenia gravis and COVID-19: Four case reports. J. Neurol. Neurosurg. Psychiatry 2020, 91, 1124–1125. [Google Scholar] [CrossRef]
  143. Meriggioli, M.N.; Sanders, D.B. Autoimmune myasthenia gravis: Emerging clinical and biological heterogeneity. Lancet Neurol. 2009, 8, 475–490. [Google Scholar] [CrossRef] [Green Version]
  144. Tropea, M.R.; Li Puma, D.D.; Melone, M.; Gulisano, W.; Arancio, O.; Grassi, C.; Conti, F.; Puzzo, D. Genetic deletion of α7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer’s disease-like pathology. Prog. Neurobiol. 2021, 206, 102154. [Google Scholar] [CrossRef]
  145. von Wrede, R.; Rings, T.; Schach, S.; Helmstaedter, C.; Lehnertz, K. Transcutaneous auricular vagus nerve stimulation induces stabilizing modifications in large-scale functional brain networks: Towards understanding the effects of taVNS in subjects with epilepsy. Sci. Rep. 2021, 11, 7906. [Google Scholar] [CrossRef]
  146. Ahmed, U.; Graf, J.F.; Daytz, A.; Yaipen, O.; Mughrabi, I.; Jayaprakash, N.; Cotero, V.; Morton, C.; Deutschman, C.S.; Zanos, S.; et al. Ultrasound Neuromodulation of the Spleen Has Time-Dependent Anti-Inflammatory Effect in a Pneumonia Model. Front. Immunol. 2022, 13, 892086. [Google Scholar] [CrossRef]
  147. Bassi, G.S.; Kanashiro, A.; Coimbra, N.C.; Terrando, N.; Maixner, W.; Ulloa, L. Anatomical and clinical implications of vagal modulation of the spleen. Neurosci. Biobehav. Rev. 2020, 112, 363–373. [Google Scholar] [CrossRef]
  148. Dhawan, S.; De Palma, G.; Willemze, R.A.; Hilbers, F.W.; Verseijden, C.; Luyer, M.D.; Nuding, S.; Wehkamp, J.; Souwer, Y.; de Jong, E.C.; et al. Acetylcholine-producing T cells in the intestine regulate antimicrobial peptide expression and microbial diversity. Am. J. Physiol. Liver Physiol. 2016, 311, G920–G933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Chang, Z.; Wang, Y.; Zhou, X.; Long, J.E. STAT3 roles in viral infection: Antiviral or proviral? Future Virol. 2018, 13, 557–574. [Google Scholar] [CrossRef] [PubMed]
  150. Grant, A.H.; Estrada, A., 3rd; Ayala-Marin, Y.M.; Alvidrez-Camacho, A.Y.; Rodriguez, G.; Robles-Escajeda, E.; Ca-dena-Medina, D.A.; Rodriguez, A.C.; Kirken, R.A. The Many Faces of JAKs and STATs Within the COVID-19 Storm. Front. Immunol. 2021, 12, 690477. [Google Scholar] [CrossRef] [PubMed]
  151. Guarnieri, T.; Abruzzo, P.M.; Bolotta, A. More than a cell biosensor: Aryl hydrocarbon receptor at the intersection of physiology and inflammation. Am. J. Physiol. Physiol. 2020, 318, C1078–C1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Giovannini-Chami, L.; Vogel, T.P.; Forbes, L.R.; Fabre, A.; Trojani, M.-C.; Leroy, S.; Antunes, O.; Vincent-Mefitiot, N.; Hiéronimus, S.; Baque-Juston, M.; et al. STAT3 gain of function: A new aetiology of severe rheumatic disease. Rheumatology 2018, 58, 365–367. [Google Scholar] [CrossRef]
  153. Milner, J.D.; Vogel, T.P.; Forbes, L.; Ma, C.A.; Stray-Pedersen, A.; Niemela, J.E.; Lyons, J.J.; Engelhardt, K.R.; Zhang, Y.; Topcagic, N.; et al. Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 2015, 125, 591–599. [Google Scholar] [CrossRef]
  154. Jägle, S.; Heeg, M.; Grün, S.; Rensing-Ehl, A.; Maccari, M.E.; Klemann, C.; Jones, N.; Lehmberg, K.; Bettoni, C.; Warnatz, K.; et al. Distinct molecular response patterns of activating STAT3 mutations associate with penetrance of lymphoproliferation and autoimmunity. Clin. Immunol. 2019, 210, 108316. [Google Scholar] [CrossRef] [PubMed]
  155. Krafft, P.R.; McBride, D.; Rolland, W.B.; Lekic, T.; Flores, J.J.; Zhang, J.H. α7 Nicotinic Acetylcholine Receptor Stimulation Attenuates Neuroinflammation through JAK2-STAT3 Activation in Murine Models of Intracerebral Hemorrhage. BioMed Res. Int. 2017, 2017, 8134653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Chatterjee, P.K.; Al-Abed, Y.; Sherry, B.; Metz, C.N. Cholinergic agonists regulate JAK2/STAT3 signaling to suppress endothelial cell activation. Am. J. Physiol. Physiol. 2009, 297, C1294–C1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Monocytes are directly innervated by the vagus nerve (VN) via alpha 7 nicotinic acetylcholine receptor (α7nAChR). Impaired ACh signaling alters the AhR/STAT3/IL-22 axis, generating a SARS-CoV-2-friendly microenvironment. ACh phosphorylates AhR and STAT3, lowering viral infection. AhR enters the nucleus, where it facilitates the transcription of IL-22, promoting the beneficial gut microbes Bifidobacterium and Lactobacillus. These probiotics, in turn, phosphorylate AhR further. Aberrant phosphorylation of the AhR/STAT3/IL-22 axis promotes viral infection, disrupting gut-barrier homeostasis.
Figure 1. Monocytes are directly innervated by the vagus nerve (VN) via alpha 7 nicotinic acetylcholine receptor (α7nAChR). Impaired ACh signaling alters the AhR/STAT3/IL-22 axis, generating a SARS-CoV-2-friendly microenvironment. ACh phosphorylates AhR and STAT3, lowering viral infection. AhR enters the nucleus, where it facilitates the transcription of IL-22, promoting the beneficial gut microbes Bifidobacterium and Lactobacillus. These probiotics, in turn, phosphorylate AhR further. Aberrant phosphorylation of the AhR/STAT3/IL-22 axis promotes viral infection, disrupting gut-barrier homeostasis.
Ijms 24 12648 g001
Figure 2. Exogenous and endogenous viruses can induce cellular senescence and syncytia formation by fusing host cells with each other. The molecular machinery of cell–cell fusion consists of Ca2+, TMEM16F, and externalized phosphatidylserine (ePS). Fusion is initiated by a fusion pore that gets larger until the cells share the cytoplasm, nuclei, and organelles. Local pH plays a major role in the initial step of pore formation.
Figure 2. Exogenous and endogenous viruses can induce cellular senescence and syncytia formation by fusing host cells with each other. The molecular machinery of cell–cell fusion consists of Ca2+, TMEM16F, and externalized phosphatidylserine (ePS). Fusion is initiated by a fusion pore that gets larger until the cells share the cytoplasm, nuclei, and organelles. Local pH plays a major role in the initial step of pore formation.
Ijms 24 12648 g002
Figure 3. The SARS-CoV-2 virus requires iron for replication. It can obtain this biometal directly from hemoglobin via open reading frame 3 and 10 (ORF3 and ORF10) or indirectly by inducing ferrosenescence. The S1 antigen blockade of ACE-2 upregulates ANG II, which increases intracellular iron by overstimulating angiotensin II type 1 receptors (AT-1Rs). The S2 antigen of SARS-CoV-2 disables p53, disrupting DNA repair.
Figure 3. The SARS-CoV-2 virus requires iron for replication. It can obtain this biometal directly from hemoglobin via open reading frame 3 and 10 (ORF3 and ORF10) or indirectly by inducing ferrosenescence. The S1 antigen blockade of ACE-2 upregulates ANG II, which increases intracellular iron by overstimulating angiotensin II type 1 receptors (AT-1Rs). The S2 antigen of SARS-CoV-2 disables p53, disrupting DNA repair.
Ijms 24 12648 g003
Figure 4. Cell–cell fusion in long COVID. The fact that mature neurons re-entering the cell cycle has been known for over two decades, however, the fact that this may be a consequence of viral infections has not been considered until the COVID-19 pandemic. Mature neurons engaging in mitosis can contribute to aneuploidy, polyploidy, and somatic mosaicism, documented by previous studies. Impaired information processing in fused neurons may explain COVID-associated neurocognitive dysfunction (CAND).
Figure 4. Cell–cell fusion in long COVID. The fact that mature neurons re-entering the cell cycle has been known for over two decades, however, the fact that this may be a consequence of viral infections has not been considered until the COVID-19 pandemic. Mature neurons engaging in mitosis can contribute to aneuploidy, polyploidy, and somatic mosaicism, documented by previous studies. Impaired information processing in fused neurons may explain COVID-associated neurocognitive dysfunction (CAND).
Ijms 24 12648 g004
Figure 5. Exogenous viruses, including SARS-CoV-2, can activate human endogenous retroviruses (HERVs) and inhibit p53, destabilizing the genome. This activates the transcription of HERV-W ENV, resulting in excessive Syncytn-1, which promotes cell–cell fusion, including neuronal fusion.
Figure 5. Exogenous viruses, including SARS-CoV-2, can activate human endogenous retroviruses (HERVs) and inhibit p53, destabilizing the genome. This activates the transcription of HERV-W ENV, resulting in excessive Syncytn-1, which promotes cell–cell fusion, including neuronal fusion.
Ijms 24 12648 g005
Figure 6. SARS-CoV-2/host protein–protein interactions, inducing endothelial senescence. Direct mechanisms include NSP6/mTORC1 and indirect ones include mitochondria, angiotensin II (ANG II), IL-17, and SIGMA-1/dopamine. Senescence activates endothelial cells (ECs) and alters the permeability of the gut barrier, enabling microbial migration outside the GI tract, a pathology likely accounting for the symptoms of long COVID.
Figure 6. SARS-CoV-2/host protein–protein interactions, inducing endothelial senescence. Direct mechanisms include NSP6/mTORC1 and indirect ones include mitochondria, angiotensin II (ANG II), IL-17, and SIGMA-1/dopamine. Senescence activates endothelial cells (ECs) and alters the permeability of the gut barrier, enabling microbial migration outside the GI tract, a pathology likely accounting for the symptoms of long COVID.
Ijms 24 12648 g006
Figure 7. taVNS activates the auricular vagus nerve, opposing the long COVID-mediated “brain fog”. The efferent vagal fibers originate in the nucleus tractus solitarius and innervate microglia/monocytes/macrophages, ECs, and IECs via α7nAChR. Upregulated ACh exerts antiviral and anti-inflammatory properties, likely clearing the virus from reservoirs.
Figure 7. taVNS activates the auricular vagus nerve, opposing the long COVID-mediated “brain fog”. The efferent vagal fibers originate in the nucleus tractus solitarius and innervate microglia/monocytes/macrophages, ECs, and IECs via α7nAChR. Upregulated ACh exerts antiviral and anti-inflammatory properties, likely clearing the virus from reservoirs.
Ijms 24 12648 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sfera, A.; Rahman, L.; Zapata-Martín del Campo, C.M.; Kozlakidis, Z. Long COVID as a Tauopathy: Of “Brain Fog” and “Fusogen Storms”. Int. J. Mol. Sci. 2023, 24, 12648. https://doi.org/10.3390/ijms241612648

AMA Style

Sfera A, Rahman L, Zapata-Martín del Campo CM, Kozlakidis Z. Long COVID as a Tauopathy: Of “Brain Fog” and “Fusogen Storms”. International Journal of Molecular Sciences. 2023; 24(16):12648. https://doi.org/10.3390/ijms241612648

Chicago/Turabian Style

Sfera, Adonis, Leah Rahman, Carlos Manuel Zapata-Martín del Campo, and Zisis Kozlakidis. 2023. "Long COVID as a Tauopathy: Of “Brain Fog” and “Fusogen Storms”" International Journal of Molecular Sciences 24, no. 16: 12648. https://doi.org/10.3390/ijms241612648

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

Article Metrics

Back to TopTop