Open access peer-reviewed chapter

COVID-19: A Catalyst for Novel Psychiatric Paradigms - Part 1

Written By

Adonis Sfera, Carolina Osorio, Jose E. Campo Maldonado, Afzaal Jafri, Aaron D. Chokka, Carlos Manuel Zapata Martín del Campo and Zisis Kozlakidis

Submitted: 25 November 2020 Reviewed: 01 March 2021 Published: 03 May 2021

DOI: 10.5772/intechopen.96940

From the Edited Volume

Biotechnology to Combat COVID-19

Edited by Megha Agrawal and Shyamasri Biswas

Chapter metrics overview

398 Chapter Downloads

View Full Metrics

Abstract

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) emerged in the late 2019 and spread rapidly throughout the world, becoming a pandemic in March 2020. It became obvious early that the prognosis of this illness is highly variable, ranging from few mild symptoms to severe complications and death, indicating that aside from the pathogen virulence, host factors contribute significantly to the overall outcome. Like SARS-CoV and Human Coronavirus NL63 (HCoV-NL63-NL63), SARS-CoV-2 enters host cells via several receptors among which angiotensin converting enzyme-2 (ACE-2) are the most studied. As this protein is widely expressed in the lungs, blood vessels, brain, kidney, testes and ovaries, the effects of this virus are widespread, affecting many body tissues and organs. Viral attachment to ACE-2 downregulates this protein, disrupting angiotensin II (ANG II) hydrolysis that in return contributes to the unchecked accumulation of this peptide. ANG II toxicity is the result of excessive activation of ANG II type 1 receptors (AT-1Rs) and N-methyl-D-aspartate NMDA receptors (NMDARs). Overstimulation of these proteins, along with the loss of angiotensin (1–7) (ANG 1–7), upregulates reactive oxygen species (ROS), inflicting end-organ damage (hit 1). However, a preexistent redox impairment may be necessary for the development of SARS-CoV-2 critical illness (hit 2). Here we propose a two-hit paradigm in which COVID-19 critical illness develops primarily in individuals with preexistent antioxidant dysfunction. Several observational studies are in line with the two hit model as they have associated poor COVID-19 prognosis with the hereditary antioxidant defects. Moreover, the SARS-CoV-2 interactome reveals that viral antigen NSP5 directly inhibits the synthesis of glutathione peroxidase (GPX), an antioxidant enzyme that along with glucose-6-phosphate dehydrogenase (G6PD) protect the body from oxidative damage. Indeed, individuals with G6PD deficiency have less favorable COVID-19 outcomes compared to the general population.

Keywords

  • Sars-CoV-2
  • antiviral psychotropic drugs
  • glucose-6-phosphate dehydrogenase
  • glutathione peroxidase
  • endocytic pathway
  • calmodulin

1. Introduction

The COVID-19 pandemic has altered many aspects of daily life, contributing to the higher incidence of psychiatric conditions, including depression, anxiety, posttraumatic stress disorder (PTSD) and substance use [1, 2, 3, 4, 5, 6, 7, 8]. In addition, as SARS-CoV-2 is a neurotropic virus, delirium, cognitive impairment and psychosis were demonstrated in up to 40% of infected patients [9, 10, 11]. Moreover, like the previous pandemics, COVID-19 may be followed by delayed or even next-generation neuropsychiatric sequelae [12, 13, 14]. For example, the offspring of women pregnant during the 1918 influenza pandemic achieved lower education, socioeconomic status, and income as adults, indicating hidden and long-lasting effects [15] (Figure 1).

Figure 1.

The two-hit paradigm: Excessive angiotensin II (ANG II) and loss of angiotensin (1–7) (ANG 1–7) generate oxidative stress both directly and indirectly (via ANG II-AT-1R and ANG II-NMDAR axes). COVID-19 critical illness is triggered when a preexistent redox dysfunction (second hit) is present.

Advertisement

2. COVID-19 and psychotropic drugs

Several psychotropic drugs have been associated with antiviral and antitumor properties, suggesting that they may lower the severity of COVID-19 critical illness [16]. For example, imipramine, clomipramine and the phenothiazine class of drugs have demonstrated efficacy against other viruses, including Ebola, Dengue and West Nile [17, 18, 19, 20]. In addition, thioridazine, another phenothiazine, was found to slow the progression of lung cancers, probably by enhancing antitumor immunity [21]. Other antipsychotics evidenced some beneficial effects in patients with glioblastoma and pancreatic cancer, suggesting immuno-oncological properties [22].

The recently published SARS-CoV-2/host protein–protein interaction and phosphorylation studies demonstrated viral interference with several pathways previously implicated in psychiatric disorders and targeted by psychotropic drugs [23, 24]. For example, upon binding to ACE-2, the SARS-CoV-2 virus ingresses host cells via the endocytic pathway (EP), a vesicular system inhibited by chlorpromazine (CPZ) and linked to schizophrenia and neurodegenerative disorders [25, 26]. Indeed, several antipsychotic drugs were found to interact with both the EP and extracellular vesicles (EVs), demonstrating previously unknown mechanisms of action [27, 28]. Other pathways involved in both the SARS-CoV-2 infection and psychiatric illness include autophagy, redox and calmodulin systems, connecting the virus to neuropathology [23, 29, 30, 31].

Several studies have associated NMDARs with sigma-1 nonopioid receptor, a protein hijacked by the SARS-CoV-2 to enable viral entry and replication [2432]. Indeed, sigma-1 agonists, such as fluvoxamine, sertraline and the antipsychotic drug, haloperidol inhibit exploitation of sigma-1, dampening viral ingress [33, 34, 35]. In addition, fluvoxamine was found to decrease ANG II-induced cardiac hypertrophy, indicating protective effects against both the SARS-CoV-2 infection and its complication [36]. Moreover, ifenprodil, an NMDAR antagonist (and sigma-1 receptor agonist), is currently in phase III clinical trials for COVID-19, linking oxidative stress to the severity of SARS-CoV-2 infection [37] (NCT04382924).

In the immune compartment, both COVID-19 and schizophrenia were associated with dysregulated inflammatory processes and lower levels of regulatory T cells (Tregs), suggesting possible autoimmune pathology [38, 39, 40]. In contrast, antipsychotic drugs were found to upregulate Tregs, lowering autoimmune inflammation [39]. Indeed, NMDARs are abundantly expressed not only in the central nervous system (CNS) but also in the immune compartment where they regulate T-cell proliferation in response to antigens. Along these lines, NMDAR antagonists, including antipsychotic drugs upregulate Tregs, enhancing immunological tolerance that in return decreases neuroinflammation [41].

In the following sections, we take a closer look at the SARS-CoV-2 interactome, looking for pathways altered by viral infection, psychiatric disorders and the action mechanism of psychotropic drugs. In other words, learning from the virus to design better psychiatric treatments (Table 1).

SARS-CoV-2PhenothiazinesReferences
Internalization via EP endocytosisInhibit EP endocytosis[42]
Lowers autophagyAugment autophagy[43]
Augments calmodulinLower calmodulin[44]
Augments sigma-1 receptor signalingLower sigma-1 receptor signaling[45]
Lower regulatory T cells (Tregs) numberUpregulate the number of regulatory T cells (Tregs)[39]

Table 1.

Phenothiazine class of antipsychotic drugs oppose several SARS-CoV-2 actions.

Advertisement

3. The SARS-CoV-2 interactome and viral infection

SARS-CoV-2 is an enveloped, positive-sense, single-stranded, RNA virus with a genome of 30 kb, encoding for 29 viral proteins. These proteins target about 332 human molecules, some of which are also involved in psychiatric disorders and the action mechanism of psychotropic drugs [24]. The virus accesses host cells via its spike (S) glycoprotein that attaches to the cell surface receptor ACE-2 [46]. Viral binding is mediated by TMPRSS2, a human protease, that cleaves S antigen into the S1 subunit, the receptor binding site, and S2, the mediator of viral fusion with host cell membranes [47]. Upon fusion the virus is internalized through the EP pits that join the early and late endosomes, reaching the lysosomes. The later, link the EP to autophagy via autolysosomes (autophagosomes fused with lysosomes) (Figure 2).

Figure 2.

Upon receptor binding and fusion, SARS-CoV-2/ACE-2 complexes enter human cells through the endocytic pathway (EP) pits early and late endosomes that subsequently join the lysosomes. Lysosomes link the EP to autophagy as authophagosomes (that can also carry the virus) fuse with the lysosomes, engendering the autolysosomes. Viruses are released from the endoplasmic reticulum - Golgi intermediate compartment (ERGIC)(not shown) to the cell surface, either individually or packed in extracellular vesicles (EVs). Viruses connected to ACE-2 receptors that are not endocytosed are shed by ADAM17. Both endocytosis and shedding contribute to ACE-2 downregulation, a marker of COVID-19 critical illness.

The SARS-CoV-2 protein–protein interaction studies have reported that 40% of viral proteins interact with human EP, indicating that vesicular trafficking plays a crucial role in COVID-19 pathogenesis [24]. In addition, several viral proteins usurp mitochondria and autophagy, cellular systems associated with host antiviral defenses [48]. Indeed, the SARS-CoV-2 interactome revealed that the virus hijacks both the mammalian target of rapamycin complex 1 (mTORC1), the master regulator of autophagy, and the E3 ubiquitin ligases in the outer mitochondrial membrane [24].

Upon release from EP into the cytosol, the SARS-CoV-2 virus replicates and assembles in the endoplasmic reticulum - Golgi intermediate compartment (ERGIC) from which the viral progeny is released at the cell surface [49].

Advertisement

4. The SARS-CoV-2/ACE-2 attachment

Novel studies have reported that the S antigen of SARS-CoV-2 virus attaches with high affinity to ACE-2 receptors, promoting oxidative stress by several mechanisms, including ANG 1–7 downregulation, ANG II accumulation and NMDRs or AT-1Rs overstimulation (Figure 1) [49, 50, 51, 52, 53, 54].

Aside from the S antigen, several other SARS-CoV-2 proteins interact directly with the human molecules, disrupting numerous pathways, including EP, epigenome, mitochondria and autophagy (Table 2).

SARS-CoV-2 proteinsHuman proteinsReferences psychiatric pathology
NSP4, NSP8, ORF9CMitochondrial dysfunction/oxidative stress[55]
NSP2, NSP6, NSP7, NSP10, NSP13, NSP15, ORF3A, E, M, ORF8Endocytic pathway (EP)[56]
NSP6, ORF9CSigma receptors, Autophagy[57]
NSP5, NSP8, NSP13, EEpigenome[58, 59]

Table 2.

The SARS-CoV-2 non-S antigen interactions with human proteins.

Both the S antigen and non-S-induced molecular changes affect molecular pathways previously associated with schizophrenia and autism. For example, excessive NMDAR activation and externalization of phosphatidylserine (PS) on the outer leaflet of plasma membrane was documented in both COVID-19 critical illness and schizophrenia [60]. This is relevant because PS exposure has been linked to dysregulated immunosuppression and the activation of coagulation cascade, changes associated with severe COVID-19 and some psychiatric disorders [61, 62]. With the same token, NMDAR/PS exposure facilitates SARS-CoV-2 endocytosis via the EP [63, 64, 65]. Interestingly, PS externalization was associated with schizophrenia as it inhibits monoamine oxidase B (MAO-B), a dopamine (DA) metabolizing enzyme [66]. Loss of MAO-B with subsequent DA upregulation is believed to trigger psychosis, linking PS exposure to severe psychiatric conditions. Furthermore, other studies have associated normal aging with EP upregulation, likely explaining the increased risk of COVID-19 complications in elderly [67].

Advertisement

5. ACE-2 downregulation

The SARS-CoV-2 fusion with host cellular membrane occurs at the level of EP pits, structures comprised of the clathrin heavy chains and adaptor protein 2 (AP2), molecules altered by both schizophrenia and the psychotropic drugs [25, 68, 69, 70] (Figure 2).

The SARS-CoV-2/ACE-2 complexes that are not endocytosed, are shed by ADAM17, contributing to ACE-2 downregulation and increased COVID-19 severity. The exacerbation of SARS-CoV-2 infection is likely the result of virus/ACE-2 complexes dissemination throughout the body via the circulatory system, increasing infectivity (Figure 3) [71].

Figure 3.

Activation of ANG II-NMDAR axis results in intracellular calcium influx and calmodulin upregulation. Calmodulin-activated ADAM17 orchestrates the shedding of ACE-2/SARS-CoV-2 complexes, leading to ACE-2 downregulation and high infectivity by ACE-2/SARS-CoV-2 circulatory dissemination.

Novel studies have shown that oxidative stress can directly activate ADAM17, triggering ACE-2 downregulation [72, 73]. This takes place as NMDARs interacts with dopamine 1 receptors (D1Rs) activating ADAM17 to excessively cleave ACE-2 from the cell membranes [74, 75, 76]. Moreover, ADAM17 can be activated directly by viral proteins NSP6 and ORF9C interaction with sigma-1 receptors [24, 77] (Table 2). Furthermore, PS exposure at the cell surface facilitates ACE-2 downregulation, suggesting that the virus may utilize multiple mechanisms to lower this protein and enable infectivity [78].

Another novel study found that ACE-2 contains a calmodulin-binding site, implicating calcium in ADAM17 activation and COVID-19 critical illness [79, 80]. Indeed, it was established that intracellular calcium influx via NMDARs upregulates calmodulin, activating ADAM17 (Figure 3) [81, 82]. On the other hand, calmodulin antagonists, including psychotropic drugs amitriptyline, phenothiazines and melatonin, inhibit ACE-2 downregulation and the odds of COVID-19 complications [83]. In addition, recent studies found that SARS-CoV-2 could activate calcium/calmodulin-dependent protein kinase II (CAMK II), linking the virus further to excitotoxicity (excessive intracellular calcium) [23].

Taken together, ADAM17 promotes ACE-2 downregulation via oxidative stress mediated by NMDARs-upregulated intracellular calcium, mechanisms involved in schizophrenia, drug addictions and COVID-19 critical illness [83, 84, 85, 86].

Advertisement

6. COVID-19: a catalyst for novel psychiatric paradigms - part 2

6.1 The virus and madness

The connection between viruses, and psychiatric disorders has been around for many centuries. In the ancient world, Thucydides reported “total and immediate loss of memory” in the survivors of “plague of Athens”, a disease suggestive of viral encephalitis [87, 88]. In our time, MRI studies have associated herpes simplex encephalitis, a condition marked by amnesia, with specific neuroimaging markers, linking viruses to cognition [89]. In addition, novel genetic studies have demonstrated that the HK2 retrovirus, frequently detected in the genome of drug addicts, was an ancestral pathogen incorporated into human DNA [90]. Over the past century, numerous studies linked in utero or early postnatal viral infections with the development of schizophrenia and autism later in life [91]. For example, women pregnant during the 1964 rubella epidemic in the United States gave birth to offspring that frequently developed autism or schizophrenia, suggesting that other viruses, probably including COVID-19, may have similar outcomes [92, 93]. In addition, obsessive–compulsive disorder (OCD), schizophrenia, attention deficit hyperactivity disorder (ADHD) and Tourette syndrome were traced to prenatal viral infections [94]. Neurodegenerative disorders, especially Parkinson’s disease (PD), were documented to surge after prior pandemics, including the 1918 influenza, suggesting that COVID-19 may promote neurodegeneration [95]. On the positive side, the SARS-CoV-2 virus may prompt the development of novel PD therapies, including angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEi) that have demonstrated efficacy in animal models [96].

Aside from linking prenatal viral exposure to severe psychiatric illness, several new studies reported that dormant CNS viruses could also engender this pathology [97]. For example, a recent report found that compared to controls, patients with schizophrenia demonstrated higher titers of Borna disease virus (BDV) immune complexes [98]. Others have connected influenza A, varicella-zoster, herpes simplex, hepatitis C and human immunodeficiency virus with the development of serious psychiatric disorders [99].

Autoantibodies against NMDARs, demonstrated in some schizophrenia patients, were recently found to be the result of molecular mimicry between the M2 protein of influenza A virus and NMDARs [100, 101]. Indeed, several large epidemiological studies found increased prevalence of autoimmune diseases in patients with schizophrenia, indicating that autoantibodies may be the result of either molecular mimicry or virus-induced modifications in human proteins [102]. For example, the molecular resemblance between an H1N1 influenza antigen and human hypocretin molecule triggers narcolepsy as virus-induced hypocretin modification may elicit autoantibodies [103]. Along these lines, the NMDAR partial antagonist, memantine, utilized in Alzheimer’s disease (AD), was found to possess immunosuppressant properties [39, 103, 104]. Indeed, prior studies have demonstrated memantine efficacy against Trypanosoma cruzi, a disease with established autoimmune pathogenesis [105].

Untreated patients with schizophrenia were reported to be at high risk of COVID-19 complications, probably due to SARS-CoV-2-associated neuroinflammation, an established risk factor of many psychiatric disorders. On the other hand, psychotropic drugs with anti-inflammatory properties may lower the SARS-CoV-2-mediated neuroinflammation, explaining the protective effects of these agents [24, 106].

6.2 COVID-19 and acquired antioxidant defects

According to the two-hit paradigm presented here, the COVID-19 prognosis is likely determined by the status of premorbid redox reserves, especially those comprised of the antioxidant enzymes G6PD and GPX. These proteins maintain homeostasis by neutralizing ANG II-activated NADPH oxidase (NOX) [107]. NOX upregulation was documented in patients with neurodegenerative disorders, schizophrenia, and suicidal behaviors, linking CNS pathology to redox system failure [108, 109, 110].

G6PD is a potent antioxidant enzyme that lowers NOX by upregulating the synthesis of NADPH and glutathione (GSH) [111]. Conversely, G6PD deficiency was associated with hemolysis and endothelial dysfunction caused by lower GSH and increased oxidative stress [111].

We surmise that the SARS-CoV-2 virus engenders acquired deficits of G6PD and GPX via ANG II-aldosterone upregulated NOX [112] (Figure 4). When COVID-19-induced deficiency of antioxidant enzymes occurs on the background of a hereditary G6PD deficit (observed in some populations with ancestral exposure to malaria), the resultant redox failure trigger COVID-19 critical illness [113] (Figure 4).

Figure 4.

The SARS-CoV-2 virus causes oxidative stress by inhibiting both GPX (directly) and G6PD (indirectly via ANG II and aldosterone-upregulated NOX). Individuals with hereditary G6PD deficiency are at higher risk for developing COVID-19 critical illness as the loss of antioxidant enzymes is more profound and oxidative stress higher.

Several recent studies have supported this model as they established that G6PD deficient individuals, including many African Americans, are more likely to develop COVID-19 critical illness [6, 7, 114, 115, 116]. Moreover, G6PD deficiency was associated with cardiovascular disease, hypertension, liver fibrosis and iron dyshomeostasis, indicating the importance of redox balance in this pathology [117, 118, 119, 120, 121].

6.2.1 Malaria and COVID-19 prognosis

Malaria is an old enemy of mankind that throughout the past centuries exacted a heavy toll on the population of Africa and the surrounding regions. Residents of these areas have gradually developed phenotypes of plasmodium-resistant erythrocytes, including G6PD deficiency, thalassemia, and hemoglobin C, to protect against malaria [122]. Although these modified red blood cells may block plasmodial ingress, individuals with these changes are more susceptible to hemolysis and iron-mediated oxidative stress that in turn promote infections, hypertension, cancer and neuropsychiatric disorders [123, 124, 125, 126]. Indeed, both Plasmodium falciparum and the SARS-CoV-2 virus induce redox dysfunctions conducive to these pathologies.

Neuropsychiatric manifestations of malaria have been known since the ancient era however, they were more thoroughly studied only in World War I when French Army physicians encountered malaria during the campaign in Northern Greece [127, 128, 129]. More recent studies demonstrated that ROS play a major role in the pathogenesis of malaria and the CNS manifestations of this infection. For example, excessive ROS were shown to directly activate nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing-3 (NLRP3) inflammasomes, molecular structures involved in numerous pathological processes, including t psychiatric disorders [130, 131]. Interestingly, some antipsychotic drugs, including clozapine, function as NLRP3 inhibitors, indicating anti-neuroinflammatory properties [132]. The SARS-CoV-2 interactome established that viral protein OPR3a can activate NLRP3 directly, suggesting a pathway for virus-induced neuroinflammation [133, 134].

Several studies reported that Plasmodium falciparum-infected red blood cells externalize PS, a phenomenon observed in severe COVID-19 illness [135]. On the other hand, CPZ was demonstrated to bind PS, promoting eryptosis (elimination of infected red blood cells) with improvement of malaria symptoms [136, 137]. Interestingly antimalarial drugs, chloroquine and hydroxychloroquine operate by inhibiting the EP, a common mechanism of action with some antipsychotic drugs, including CPZ [138]. Since erythrocytes with externalized PS were also documented hypertension, further studies are needed to clarify the role of PS in illness and eryptosis as a possible therapeutic intervention [138, 139]. Indeed, CPZ has been utilized routinely in the emergency treatment of uncontrolled hypertension, indicating a possible role of eryptosis in addition to the well-established CPZ effects on alpha-adrenergic receptors [140].

6.2.2 Malaria exposure and the risk of COVID-19

Population groups throughout the world with exposure to malaria during the previous centuries were found to be at higher risk of hereditary G6PD deficiency and antioxidant failure. This background increases the odds not only of viral infections but also of other redox disorders, including hypertension, cancer and cardiac disease. For example, 12.2% of African American males and 4.1% of females are G6PD deficient, indicating a potentially higher risk of COVID-19 complications [141]. Indeed, novel studies found a 2.4 percent higher COVID-19 mortality in African Americans compared to Whites, Asians or Latinos [142].

Moreover, the lower GSH and nitric oxide (NO) levels in African Americans compared to other groups, places this population at higher risk of both hypertension and prostate cancer, suggesting that the SARS-CoV-2 infection may precipitate these complications [143, 144, 145, 146, 147, 148, 149]. In this regard, African Americans with COVID-19 should be routinely assessed for G6PD deficiency and supplemented with the widely available antioxidant, N-acetylcysteine [150].

Oxidative stress was demonstrated to directly trigger hypertension by resetting the CNS baroreflex, therefore the G6PD-deficient individuals could be more prone to COVID-19-related cardiovascular complications [151]. On the other hand, ARBs and ACEi lower ANG II-mediated ROS, likely averting these complications [152, 153, 154, 155, 156, 157]. Indeed, the lower utilization of ARBs and ACEi in the treatment of hypertensive African Americans may place this population at higher risk of COVID-19 critical illness [158]. Although numerous clinical trials supported the efficacy of ARBs and ACEi in African Americans, these drugs are rarely utilized in this population as an initial therapeutic options [158, 159]. This is significant as both ARBs and ACEi appear to lower COVID-19 mortality rate, probably by dampening oxidative stress-ACE-2 downregulation. For example, a novel study found that COVID-19 patients treated with ACEi or ARBs at the time of initial infection had fewer unfavorable outcomes and lower mortality rate compared to individuals unexposed to these drugs [160].

Taken together, the SARS-CoV-2-upregulated ANG II, triggers hypertension and cardiovascular disease by augmenting oxidative stress and altering the baroreceptor setting. Individuals with G6PD deficiency are at increased risk of both hypertension and COVID-19 critical illness, indicating alignment with the two-hit paradigm presented here.

Advertisement

7. Conclusion

The COVID-19 pandemic has exacerbated the disease course in many psychiatric patients as mandatory social isolation and decreased frequency of therapeutic meetings promoted fear and uncertainty in this fragile population. The restrictive measures associated with the pandemic have often led to decreased medication adherence, increased depression, anxiety and substance use disorders, often contributing to unfavorable outcomes.

On a positive note, the SARS-CoV-2 virus may be a catalyst for a better understanding of the role of viruses in the pathogenesis of psychiatric illness. As SARS-CoV-2 (and probably other viruses) utilize the molecular machinery involved in severe psychiatric disorders, the clarification of these mechanisms may help with the development of better therapies. Indeed, the EP and antioxidant enzymes may become the new psychiatric paradigms, expanding the current dopamine and serotonin models to include viruses and microbes in psychopathology.

References

  1. 1. Mathewson AC, Bishop A, Yao Y, Kemp F, Ren J, Chen H, Xu X, Berkhout B, van der Hoek L, Jones IM. Interaction of severe acute respiratory syndrome-coronavirus and NL63 coronavirus spike proteins with angiotensin converting enzyme-2. J Gen Virol. 2008 Nov;89(Pt 11):2741-2745. doi: 10.1099/vir.0.2008/003962-0. PMID: 18931070; PMCID: PMC2886958.
  2. 2. Verdecchia P, Cavallini C, Spanevello A, Angeli F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur J Intern Med. 2020;76:14-20. doi:10.1016/j.ejim.2020.04.037
  3. 3. Sfera A, Osorio C, Jafri N, Diaz EL, Campo Maldonado JE. Intoxication With Endogenous Angiotensin II: A COVID-19 Hypothesis. Front Immunol. 2020;11:1472. Published 2020 Jun 19. doi:10.3389/fimmu.2020.01472
  4. 4. Sasi, Sreethish, et al. "A case of COVID-19 in a patient with asymptomatic hemoglobin D thalassemia and glucose-6-phosphate dehydrogenase deficiency." The American Journal of Case Reports 21 (2020): e925788-e925781.
  5. 5. Al-Aamri, Maryam Ali, et al. "A saudi G6PD deficient girl died with pediatric multisystem inflammatory syndrome-COVID-19." MedRxiv (2020).
  6. 6. Vick, Dan J. "Glucose-6-phosphate dehydrogenase deficiency and COVID-19 infection." Mayo Clinic Proceedings. Vol. 95. No. 8. Elsevier, 2020.
  7. 7. Taylor EW, Radding W. Understanding Selenium and Glutathione as Antiviral Factors in COVID-19: Does the Viral Mpro Protease Target Host Selenoproteins and Glutathione Synthesis?. Front Nutr. 2020;7:143. Published 2020 Sep 2. doi:10.3389/fnut.2020.00143
  8. 8. Simon NM, Saxe GN, Marmar CR. Mental Health Disorders Related to COVID-19-Related Deaths. JAMA. 2020 Oct 20;324(15):1493-1494. doi: 10.1001/jama.2020.19632. PMID: 33044510.
  9. 9. Parra A, Juanes A, Losada CP, et al. Psychotic symptoms in COVID-19 patients. A retrospective descriptive study. Psychiatry Res. 2020;291:113254. doi:10.1016/j.psychres.2020.113254
  10. 10. Rogers JP, Chesney E, Oliver D, et al. Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: a systematic review and meta-analysis with comparison to the COVID-19 pandemic. Lancet Psychiatry. 2020;7(7):611-627. doi:10.1016/S2215-0366(20)30203-0
  11. 11. Banerjee D, Viswanath B. Neuropsychiatric manifestations of COVID-19 and possible pathogenic mechanisms: Insights from other coronaviruses. Asian J Psychiatr. 2020;54:102350. doi:10.1016/j.ajp.2020.102350
  12. 12. Saunders-Hastings PR, Krewski D. Reviewing the History of Pandemic Influenza: Understanding Patterns of Emergence and Transmission. Pathogens. 2016 Dec 6;5(4):66. doi: 10.3390/pathogens5040066. PMID: 27929449; PMCID: PMC5198166.
  13. 13. Manjunatha N, Math SB, Kulkarni GB, Chaturvedi SK. The neuropsychiatric aspects of influenza/swine flu: A selective review. Ind Psychiatry J. 2011;20(2):83-90. doi:10.4103/0972-6748.102479
  14. 14. Steinman G. COVID-19 and autism. Med Hypotheses. 2020;142:109797. doi:10.1016/j.mehy.2020.109797
  15. 15. Fletcher JM. The effects of in utero exposure to the 1918 influenza pandemic on family formation. Econ Hum Biol. 2018;30:59-68. doi:10.1016/j.ehb.2018.06.004
  16. 16. Plaze M, Attali D, Petit AC, et al. Repurposing chlorpromazine to treat COVID-19: The reCoVery study. Encephale. 2020;46(3):169-172. doi:10.1016/j.encep.2020.05.006
  17. 17. Zhao Y, Ren J, Fry EE, Xiao J, Townsend AR, Stuart DI. Structures of Ebola Virus Glycoprotein Complexes with Tricyclic Antidepressant and Antipsychotic Drugs. J Med Chem. 2018 Jun 14;61(11):4938-4945. doi: 10.1021/acs.jmedchem.8b00350. Epub 2018 May 21. PMID: 29741894.
  18. 18. Bocci G, Bradfute SB, Ye C, Garcia MJ, Parvathareddy J, Reichard W, et al. Virtual and In Vitro Antiviral Screening Revive Therapeutic Drugs for COVID-19. ACS Pharmacology & Translational Science(2020). DOI: 10.1021/acsptsci.0c00131
  19. 19. Otręba, Michał, Leon Kośmider, and Anna Rzepecka-Stojko. "Antiviral activity of chlorpromazine, fluphenazine, perphenazine, prochlorperazine, and thioridazine towards RNA-viruses. A review." European journal of pharmacology 887 (2020): 173553.
  20. 20. Wang, Yancui, et al. "Inactivation of Zika virus in plasma and derivatives by four different methods." Journal of medical virology 91.12 (2019): 2059-2065.
  21. 21. Shen J, Ma B, Zhang X, et al. Thioridazine has potent antitumor effects on lung cancer stem-like cells. Oncol Lett. 2017;13(3):1563-1568. doi:10.3892/ol.2017.5651
  22. 22. Tuan NM, Lee CH. Penfluridol as a Candidate of Drug Repurposing for Anticancer Agent. Molecules. 2019;24(20):3659. Published 2019 Oct 11. doi:10.3390/molecules24203659
  23. 23. Bouhaddou M, Memon D, Meyer B, et al. The Global Phosphorylation Landscape of SARS-CoV-2 Infection. Cell. 2020;182(3):685-712.e19. doi:10.1016/j.cell.2020.06.034
  24. 24. Gordon DE, Jang GM, Bouhaddou M, Xu J, Obernier K, White KM, O'Meara MJ, Rezelj VV, et al. SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature. 2020 Jul;583(7816):459-468. doi: 10.1038/s41586-020-2286-9. Epub 2020 Apr 30. PMID: 32353859; PMCID: PMC7431030.
  25. 25. Schubert KO, Föcking M, Prehn JH, Cotter DR. Hypothesis review: are clathrin-mediated endocytosis and clathrin-dependent membrane and protein trafficking core pathophysiological processes in schizophrenia and bipolar disorder? Mol Psychiatry. 2012 Jul;17(7):669-681. doi: 10.1038/mp.2011.123. Epub 2011 Oct 11. PMID: 21986877.
  26. 26. Wu F, Yao PJ. Clathrin-mediated endocytosis and Alzheimer's disease: an update. Ageing Res Rev. 2009 Jul;8(3):147-149. doi: 10.1016/j.arr.2009.03.002. Epub 2009 Mar 21. PMID: 19491039.
  27. 27. Kano SI, Dohi E, Rose IVL. Extracellular Vesicles for Research on Psychiatric Disorders. Schizophr Bull. 2019;45(1):7-16. doi:10.1093/schbul/sby127
  28. 28. Daniel JA, Chau N, Abdel-Hamid MK, Hu L, von Kleist L, Whiting A, Krishnan S, Maamary P, Joseph SR, Simpson F, Haucke V, McCluskey A, Robinson PJ. Phenothiazine-derived antipsychotic drugs inhibit dynamin and clathrin-mediated endocytosis. Traffic. 2015 Jun;16(6):635-654. doi: 10.1111/tra.12272. Epub 2015 Apr 9. PMID: 25693808.
  29. 29. Vucicevic L, Misirkic-Marjanovic M, Harhaji-Trajkovic L, Maric N, Trajkovic V. Mechanisms and therapeutic significance of autophagy modulation by antipsychotic drugs. Cell Stress. 2018;2(11):282-291. Published 2018 Oct 25. doi:10.15698/cst2018.11.161
  30. 30. Novak G, Seeman P, Tallerico T. Increased expression of calcium/calmodulin-dependent protein kinase IIbeta in frontal cortex in schizophrenia and depression. Synapse. 2006 Jan;59(1):61-68. doi: 10.1002/syn.20211. PMID: 16247765.
  31. 31. Yao JK, Keshavan MS. Antioxidants, redox signaling, and pathophysiology in schizophrenia: an integrative view. Antioxid Redox Signal. 2011;15(7):2011-2035. doi:10.1089/ars.2010.3603
  32. 32. Pabba M, Sibille E. Sigma-1 and N-Methyl-d-Aspartate Receptors: A Partnership with Beneficial Outcomes. Mol Neuropsychiatry. 2015;1(1):47-51. doi:10.1159/000376549
  33. 33. Friesland M, Mingorance L, Chung J, Chisari FV, Gastaminza P. Sigma-1 receptor regulates early steps of viral RNA replication at the onset of hepatitis C virus infection. J Virol. 2013;87(11):6377-6390. doi:10.1128/JVI.03557-12
  34. 34. Cobos EJ, Entrena JM, Nieto FR, Cendán CM, Del Pozo E. Pharmacology and therapeutic potential of sigma(1) receptor ligands. Curr Neuropharmacol. 2008;6(4):344-366. doi:10.2174/157015908787386113
  35. 35. Lee IT, Chen S, Schetz JA. An unambiguous assay for the cloned human sigma1 receptor reveals high affinity interactions with dopamine D4 receptor selective compounds and a distinct structure-affinity relationship for butyrophenones. Eur J Pharmacol. 2008;578(2-3):123-136. doi:10.1016/j.ejphar.2007.09.020
  36. 36. Tagashira H, Bhuiyan S, Shioda N, Hasegawa H, Kanai H, Fukunaga K. σ1-receptor stimulation with fluvoxamine ameliorates transverse aortic constriction-induced myocardial hypertrophy and dysfunction in mice, Am J Physiol, 2010, vol. 299 (pg. H1535-H1545)
  37. 37. Hashimoto K. Activation of sigma-1 receptor chaperone in the treatment of neuropsychiatric diseases and its clinical implication. J Pharmacol Sci. 2015 Jan;127(1):6-9. doi: 10.1016/j.jphs.2014.11.010. Epub 2014 Dec 4. PMID: 25704012.
  38. 38. Gladstone DE, Kim BS, Mooney K, Karaba AH, D'Alessio FR. Regulatory T Cells for Treating Patients With COVID-19 and Acute Respiratory Distress Syndrome: Two Case Reports. Ann Intern Med. 2020 Jul 6:L20-0681. doi: 10.7326/L20-0681. Epub ahead of print. PMID: 32628535; PMCID: PMC7370819.
  39. 39. Kelly DL, Li X, Kilday C, Feldman S, Clark S, Liu F, Buchanan RW, Tonelli LH. Increased circulating regulatory T cells in medicated people with schizophrenia. Psychiatry Res. 2018 Nov;269:517-523. doi: 10.1016/j.psychres.2018.09.006. Epub 2018 Sep 5. PMID: 30195746; PMCID: PMC6207456.
  40. 40. May M, Beauchemin M, Vary C, Barlow D, Houseknecht KL. The antipsychotic medication, risperidone, causes global immunosuppression in healthy mice. PLoS One. 2019;14(6):e0218937. Published 2019 Jun 26. doi:10.1371/journal.pone.0218937
  41. 41. Kahlfuß S, Simma N, Mankiewicz J, et al. Immunosuppression by N-methyl-D-aspartate receptor antagonists is mediated through inhibition of Kv1.3 and KCa3.1 channels in T cells. Mol Cell Biol. 2014;34(5):820-831. doi:10.1128/MCB.01273-13
  42. 42. Negreira-Caamaño M, Piqueras-Flores J, Martínez-DelRio J, et al. Impact of Treatment with Renin-Angiotensin System Inhibitors on Clinical Outcomes in Hypertensive Patients Hospitalized with COVID-19 [published online ahead of print, 2020 Sep 19]. High Blood Press Cardiovasc Prev. 2020;1-8. doi:10.1007/s40292-020-00409
  43. 43. Dutta D, Donaldson JG. Search for inhibitors of endocytosis:Intended specificity and unintended consequences. Cell Logist. 2012;2 (4):203-208. doi:10.4161/cl.23967
  44. 44. Vucicevic L, Misirkic-Marjanovic M, Harhaji-Trajkovic L, Maric N, Trajkovic V. Mechanisms and therapeutic significance of autophagy modulation by antipsychotic drugs. Cell Stress. 2018;2(11):282-291. Published 2018 Oct 25. doi:10.15698/cst2018.11.161
  45. 45. Prozialeck WC, Weiss B. Inhibition of calmodulin by phenothiazines and related drugs: structure–activity relationships. J Pharmacol Exp Ther. 1982 Sep;222 (3):509-516. PMID: 6286920.
  46. 46. Zhou, P., Yang, XL., Wang, XG. et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579, 270-273 (2020). https://doi.org/10.1038/s41586-020-2012-7
  47. 47. Mahendran ASK, Lim YS, Fang CM, Loh HS, Le CF. The Potential of Antiviral Peptides as COVID-19 Therapeutics. Front Pharmacol. 2020;11:575444. Published 2020 Sep 15. doi:10.3389/fphar.2020.575444
  48. 48. Carmona-Gutierrez D, Bauer MA, Zimmermann A, et al. Digesting the crisis: autophagy and coronaviruses. Microb Cell. 2020;7(5):119-128. Published 2020 May 4. doi:10.15698/mic2020.05.715
  49. 49. Hassanpour M, Rezaie J, Nouri M, Panahi Y. The role of extracellular vesicles in COVID-19 virus infection [published online ahead of print, 2020 Jun 13]. Infect Genet Evol. 2020;85:104422. doi:10.1016/j.meegid.2020.104422
  50. 50. Xu J, Sriramula S, Lazartigues E. Excessive Glutamate Stimulation Impairs ACE2 Activity Through ADAM17-Mediated Shedding in Cultured Cortical Neurons. Cellular and Molecular Neurobiology. 2018 Aug;38(6):1235-1243. DOI: 10.1007/s10571-018-0591-8.
  51. 51. Zhang F, Liu C, Wang L, Cao X, Wang YY, Yang JK. Antioxidant effect of angiotensin (1-7) in the protection of pancreatic β cell function. Mol Med Rep. 2016 Sep;14(3):1963-9. doi: 10.3892/mmr.2016.5514. Epub 2016 Jul 13. PMID: 27430410; PMCID: PMC4991744.
  52. 52. Sommer A, Kordowski F, Büch J, Maretzky T, Evers A, Andrä J, Düsterhöft S, Michalek M, Lorenzen I, Somasundaram P, Tholey A, Sönnichsen FD, Kunzelmann K, Heinbockel L, Nehls C, Gutsmann T, Grötzinger J, Bhakdi S, Reiss K. Phosphatidylserine exposure is required for ADAM17 sheddase function. Nat Commun. 2016 May 10;7:11523. doi: 10.1038/ncomms11523. PMID: 27161080; PMCID: PMC4866515.
  53. 53. Doumas M, Patoulias D, Katsimardou A, Stavropoulos K, Imprialos K, Karagiannis A. COVID19 and increased mortality in African Americans: socioeconomic differences or does the renin angiotensin system also contribute? [published online ahead of print, 2020 Jul 15]. J Hum Hypertens. 2020;1-4. doi:10.1038/s41371-020-0380-y
  54. 54. Welch WJ. Angiotensin II-dependent superoxide: effects on hypertension and vascular dysfunction. Hypertension. 2008;52(1):51-56. doi:10.1161/HYPERTENSIONAHA.107.090472
  55. 55. Krutetskaya ZI, Melnitskaya AV, Antonov VG, Nozdrachev AD. Sigma-1 Receptor Antagonists Haloperidol and Chlorpromazine Modulate the Effect of Glutoxim on Na + Transport in Frog Skin. Dokl Biochem Biophys. 2019 May;484(1):63-65. doi: 10.1134/S1607672919010186. Epub 2019 Apr
  56. 56. Shao L, Martin MV, Watson SJ, et al. Mitochondrial involvement in psychiatric disorders [published correction appears in Ann Med. 2011 Jun;43 (4):329]. Ann Med. 2008;40 (4):281-295. doi:10.1080/07853890801923753
  57. 57. Ryder PV, Faundez V. Schizophrenia: the “BLOC” may be in the endosomes. Sci Signal. 2009;2(93):pe66. Published 2009 Oct 20. doi:10.1126/scisignal.293pe66
  58. 58. Tsai SY, Pokrass MJ, Klauer NR, De Credico NE, Su TP. sigma-1 receptor chaperones in neurodegenerative and psychiatric disorders. Expert Opin Ther Targets. 2014;18(12):1461-1476. doi:10.1517/14728222.2014.97
  59. 59. Pishva E, Creese B, Smith AR, Viechtbauer W, Proitsi P, van den Hove DLA, Ballard C, Mill J, Lunnon K. Psychosisassociated DNA methylomic variation in Alzheimer’s disease cortex. Neurobiol Aging. 2020 May;89:83-88. doi: 10.1016/j.neurobiolaging.2020.01.001. Epub 2020 Jan 8. PMID: 32007278.
  60. 60. Gasull T, Sarri E, DeGregorio-Rocasolano N, Trullas R. NMDA receptor overactivation inhibits phospholipid synthesis by decreasing choline-ethanolamine phosphotransferase activity. J Neurosci. 2003 May 15;23(10):4100-7. doi: 10.1523/JNEUROSCI.23-10-04100.2003. PMID: 12764097; PMCID: PMC6741076.
  61. 61. Hoirisch-Clapauch S, Amaral OB, Mezzasalma MA, Panizzutti R, Nardi AE. Dysfunction in the coagulation system and schizophrenia. Transl Psychiatry. 2016;6(1):e704. Published 2016 Jan 5. doi:10.1038/tp.2015.204
  62. 62. Jayarangaiah A, Kariyanna PT, Chen X, Jayarangaiah A, Kumar A. COVID-19-Associated Coagulopathy: An Exacerbated Immunothrombosis Response. Clin Appl Thromb Hemost. 2020;26:1076029620943293. doi:10.1177/1076029620943293
  63. 63. Glebov, O.O. Tonic NMDA receptor signalling shapes endosomal organisation in mammalian cells. Sci Rep 10, 9315 (2020). https://doi.org/10.1038/s41598-020-66071-0
  64. 64. Sicari D, Chatziioannou A, Koutsandreas T, Sitia R, Chevet E. Role of the early secretory pathway in SARS-CoV-2 infection [published correction appears in J Cell Biol. 2020 Sep 7;219(9):]. J Cell Biol. 2020;219(9):e202006005. doi:10.1083/jcb.202006005
  65. 65. Hasanagic S, Serdarevic F. Potential role of memantine in the prevention and treatment of COVID-19: its antagonism of nicotinic acetylcholine receptors and beyond. Eur Respir J. 2020;56(2):2001610. Published 2020 Aug 13. doi:10.1183/13993003.01610-2020
  66. 66. Tachiki KH, Buckman TD, Eiduson S, Kling AS, Hullett J. Phosphatidylserine inhibition of monoamine oxidase in platelets of schizophrenics. Biol Psychiatry. 1986 Jan;21(1):59-68. doi: 10.1016/0006-3223(86)90008-9. PMID: 3942801.
  67. 67. Alsaqati M, Thomas RS, Kidd EJ. Proteins Involved in Endocytosis Are Upregulated by Ageing in the Normal Human Brain: Implications for the Development of Alzheimer's Disease. J Gerontol A Biol Sci Med Sci. 2018 Mar 2;73(3):289-298. doi: 10.1093/gerona/glx135. PMID: 28655199.
  68. 68. Bayati A, Kumar R, Francis V, McPherson PS. SARS-CoV-2 uses clathrin-mediated endocytosis to gain access into cells. BioRxIv (2020) doi: https://doi.org/10.1101/2020.07.13.201509
  69. 69. Garcia MD, Formoso K, Aparicio GI, Frasch ACC, Scorticati C. The Membrane Glycoprotein M6a Endocytic/Recycling Pathway Involves Clathrin-Mediated Endocytosis and Affects Neuronal Synapses. Front Mol Neurosci. 2017;10:296. Published 2017 Sep 20. doi:10.3389/fnmol.2017.00296
  70. 70. Sharp SI, Hu Y, Weymer JF, et al. The effect of clozapine on mRNA expression for genes encoding G protein-coupled receptors and the protein components of clathrin-mediated endocytosis. Psychiatr Genet. 2013;23(4):153-162. doi:10.1097/YPG.0b013e32835fe51d
  71. 71. Heurich A, Hofmann-Winkler H, Gierer S, Liepold T, Jahn O, Pöhlmann S. TMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein. J Virol. 2014 Jan;88(2):1293-307. doi: 10.1128/JVI.02202-13. Epub 2013 Nov 13. PMID: 24227843; PMCID: PMC3911672.
  72. 72. Pagliaro P, Penna C. ACE/ACE2 Ratio: A Key Also in 2019 Coronavirus Disease (Covid-19)?. Front Med (Lausanne). 2020;7:335. Published 2020 Jun 18. doi:10.3389/fmed.2020.00335
  73. 73. Brill A, Chauhan AK, Canault M, Walsh MT, Bergmeier W, Wagner DD. Oxidative stress activates ADAM17/TACE and induces its target receptor shedding in platelets in a p38-dependent fashion. Cardiovasc Res. 2009;84(1):137-144. doi:10.1093/cvr/cvp176
  74. 74. Chen G, Greengard P, Yan Z. Potentiation of NMDA receptor currents by dopamine D1 receptors in prefrontal cortex. Proc Natl Acad Sci U S A. 2004;101(8):2596-2600. doi:10.1073/pnas.0308618100
  75. 75. Ladepeche L, Dupuis JP, Bouchet D, et al. Single-molecule imaging of the functional crosstalk between surface NMDA and dopamine D1 receptors. Proc Natl Acad Sci U S A. 2013;110(44):18005-18010. doi:10.1073/pnas.1310145110
  76. 76. Pei L, Lee FJ, Moszczynska A, Vukusic B, Liu F. Regulation of dopamine D1 receptor function by physical interaction with the NMDA receptors. J Neurosci. 2004;24(5):1149-1158. doi:10.1523/JNEUROSCI.3922-03.2004
  77. 77. Li J, Liu B, Gao X, et al. Overexpression of sigma-1 receptor inhibits ADAM10 and ADAM17 mediated shedding in vitro [published correction appears in Protein Cell. 2012 May;3(5):400]. Protein Cell. 2012;3(2):153-159. doi:10.1007/s13238-012-2006-9
  78. 78. Wang G, Coleman CG, Chan J, et al. Angiotensin II slow-pressor hypertension enhances NMDA currents and NOX2-dependent superoxide production in hypothalamic paraventricular neurons. Am J Physiol Regul Integr Comp Physiol. 2013;304(12):R1096-R1106. doi:10.1152/ajpregu.00367.2012
  79. 79. Lai ZW, Lew RA, Yarski MA, Mu FT, Andrews RK, Smith AI. The identification of a calmodulin-binding domain within the cytoplasmic tail of angiotensin-converting enzyme-2. Endocrinology. 2009;150(5):2376-2381. doi:10.1210/en.2008-1274
  80. 80. Lambert DW, Clarke NE, Hooper NM, Turner AJ. Calmodulin interacts with angiotensin-converting enzyme-2 (ACE2) and inhibits shedding of its ectodomain. FEBS Lett. 2008;582(2):385-390. doi:10.1016/j.febslet.2007.11.085
  81. 81. Ermak G, Davies KJ. Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol. 2002 Feb;38(10):713-721. doi: 10.1016/s0161-5890(01)00108-0. PMID: 11841831.
  82. 82. Ataman ZA, Gakhar L, Sorensen BR, Hell JW, Shea MA. The NMDA receptor NR1 C1 region bound to calmodulin: structural insights into functional differences between homologous domains. Structure. 2007;15(12):1603-1617. doi:10.1016/j.str.2007.10.012
  83. 83. Ragia G, Manolopoulos VG. Inhibition of SARS-CoV-2 entry through the ACE2/TMPRSS2 pathway: a promising approach for uncovering early COVID-19 drug therapies [published online ahead of print, 2020 Jul 21]. Eur J Clin Pharmacol. 2020;1-8. doi:10.1007/s00228-020-02963-4
  84. 84. Novak G, Seeman P, Tallerico T. Schizophrenia: elevated mRNA for calcium-calmodulin-dependent protein kinase IIbeta in frontal cortex. Brain research. Molecular Brain Research. 2000 Oct;82(1-2):95-100. DOI: 10.1016/s0169-328x(00)00188-1.
  85. 85. Soto-Vega E, Meza I, Ramírez-Rodríguez G, Benitez-King G. Melatonin stimulates calmodulin phosphorylation by protein kinase C. J Pineal Res. 2004 Sep;37(2):98-106. doi: 10.1111/j.1600-079X.2004.00141.x. PMID: 15298668.
  86. 86. Wei Y, Wang R, Teng J. Inhibition of Calcium/Calmodulin-Dependent Protein Kinase IIα Suppresses Oxidative Stress in Cerebral Ischemic Rats Through Targeting Glucose 6-Phosphate Dehydrogenase. Neurochem Res. 2019 Jul;44(7):1613-1620. doi: 10.1007/s11064-019-02785-6. Epub 2019 Mar 27. PMID: 30919283.
  87. 87. Mujtaba S, He Y, Zeng L, Yan S, Plotnikova O, Sachchidanand, Sanchez R, Zeleznik-Le NJ, Ronai Z, Zhou MM. Structural mechanism of the bromodomain of the coactivator CBP in p53 transcriptional activation. Mol Cell. 2004 Jan 30;13(2):251-263. doi: 10.1016/s1097-2765(03)00528-8. PMID: 14759370.
  88. 88. Cunha BA. The cause of the plague of Athens: plague, typhoid, typhus, smallpox, or measles? Infectious Disease Clinics of North America. 2004 Mar;18(1):29-43. DOI: 10.1016/s0891-5520(03)00100-4.
  89. 89. Kapur N, Barker S, Burrows EH, Ellison D, Brice J, Illis LS, Scholey K, Colbourn C, Wilson B, Loates M. Herpes simplex encephalitis: long term magnetic resonance imaging and neuropsychological profile. J Neurol Neurosurg Psychiatry. 1994 Nov;57(11):1334-1342. doi: 10.1136/jnnp.57.11.1334. PMID:
  90. 90. Karamitros T, Hurst T, Marchi E, Karamichali E, Georgopoulou U, Mentis A, Riepsaame J, Lin A, Paraskevis D, Hatzakis A, McLauchlan J, Katzourakis A, Magiorkinis G. Human Endogenous Retrovirus-K HML-2 integration within RASGRF2 is associated with intravenous drug abuse and modulates transcription in a cell-line model. Proc Natl Acad Sci U S A. 2018 Oct 9;115(41):10434-10439. doi: 10.1073/pnas.1811940115. Epub 2018 Sep 24. PMID: 30249655; PMCID: PMC6187174.
  91. 91. McGrath JJ, Pemberton MR, Welham JL, Murray RM. Schizophrenia and the influenza epidemics of 1954, 1957 and 1959: a southern hemisphere study. Schizophr Res. 1994 Dec;14(1):1-8. doi: 10.1016/0920-9964(94)90002-7. PMID: 7893616.
  92. 92. Hutton J. Does Rubella Cause Autism: A 2015 Reappraisal?. Front Hum Neurosci. 2016;10:25. Published 2016 Feb 1. doi:10.3389/fnhum.2016.00025
  93. 93. Brown AS, Begg MD, Gravenstein S, Schaefer CA, Wyatt RJ, Bresnahan M, Babulas VP, Susser ES. Serologic evidence of prenatal influenza in the etiology of schizophrenia. Arch Gen Psychiatry. 2004 Aug;61(8):774-780. doi: 10.1001/archpsyc.61.8.774. PMID: 15289276.
  94. 94. Khandaker GM, Zimbron J, Dalman C, Lewis G, Jones PB. Childhood infection and adult schizophrenia: a meta-analysis of population-based studies. Schizophr Res. 2012;139(1-3):161-168. doi:10.1016/j.schres.2012.05.023
  95. 95. Henry J, Smeyne RJ, Jang H, Miller B, Okun MS. Parkinsonism and neurological manifestations of influenza throughout the 20th and 21st centuries. Parkinsonism Relat Disord. 2010;16(9):566-571. doi:10.1016/j.parkreldis.2010.06.012
  96. 96. Perez-Lloret S, Otero-Losada M, Toblli JE, Capani F. Renin-angiotensin system as a potential target for new therapeutic approaches in Parkinson's disease. Expert Opin Investig Drugs. 2017 Oct;26(10):1163-1173. doi: 10.1080/13543784.2017.1371133. Epub 2017 Aug 29. PMID: 28836869.
  97. 97. Breier A. 39. VIRUSES AND SCHIZOPHRENIA: IMPLICATIONS FOR PATHOPHYSIOLOGY AND TREATMENT. Schizophr Bull. 2018;44(Suppl 1):S61-S62. doi:10.1093/schbul/sby014.158
  98. 98. Zaliunaite V, Steibliene V, Bode L, Podlipskyte A, Bunevicius R, Ludwig H. Primary psychosis and Borna disease virus infection in Lithuania: a case control study. BMC Psychiatry. 2016;16(1):369. Published 2016 Nov 3. doi:10.1186/s12888-016-1087-z
  99. 99. Coughlin SS. Anxiety and Depression: Linkages with Viral Diseases. Public Health Rev. 2012;34(2):7. doi:10.1007/BF03391675
  100. 100. Kępińska AP, Iyegbe CO, Vernon AC, Yolken R, Murray RM, Pollak TA. Schizophrenia and Influenza at the Centenary of the 1918-1919 Spanish Influenza Pandemic: Mechanisms of Psychosis Risk. Front Psychiatry. 2020;11:72. Published 2020 Feb 26. doi:10.3389/fpsyt.2020.00072
  101. 101. Blanpied TA, Clarke RJ, Johnson JW. Amantadine inhibits NMDA receptors by accelerating channel closure during channel block. J Neurosci. 2005 Mar 30;25(13):3312-22. doi: 10.1523/JNEUROSCI.4262-04.2005. PMID: 15800186; PMCID: PMC6724906.
  102. 102. Eaton WW, Byrne M, Ewald H, Mors O, Chen CY, Agerbo E, Mortensen PB. Association of schizophrenia and autoimmune diseases: linkage of Danish national registers. Am J Psychiatry. 2006 Mar;163(3):521-528. doi: 10.1176/appi.ajp.163.3.521. PMID: 16513876.
  103. 103. Luo G, Ambati A, Lin L, Bonvalet M, Partinen M, Ji X, Maecker HT, Mignot EJ. Autoimmunity to hypocretin and molecular mimicry to flu in type 1 narcolepsy. Proc Natl Acad Sci U S A. 2018 Dec 26;115(52):E12323-E12332. doi: 10.1073/pnas.1818150116. Epub 2018 Dec 12. PMID: 30541895; PMCID: PMC6310865.
  104. 104. Rejdak K, Grieb P. Adamantanes might be protective from COVID-19 in patients with neurological diseases: multiple sclerosis, parkinsonism and cognitive impairment. Multiple Sclerosis and Related Disorders. 2020;42:102163 10.1016/j.msard.2020.102163
  105. 105. Damasceno FS, Barisón MJ, Pral EM, Paes LS, Silber AM. Memantine, an antagonist of the NMDA glutamate receptor, affects cell proliferation, differentiation and the intracellular cycle and induces apoptosis in Trypanosoma cruzi. PLoS Negl Trop Dis. 2014;8(2):e2717. Published 2014 Feb 27. doi:10.1371/journal.pntd.0002717
  106. 106. Kozloff N, Mulsant BH, Stergiopoulos V, Voineskos AN. The COVID-19 Global Pandemic: Implications for People With Schizophrenia and Related Disorders. Schizophr Bull. 2020 Jul 8;46(4):752-757. doi: 10.1093/schbul/sbaa051. PMID: 32343342; PMCID: PMC7197583.
  107. 107. Wen H, Gwathmey JK, Xie LH. Oxidative stress-mediated effects of angiotensin II in the cardiovascular system. World J Hypertens. 2012;2(4):34-44. doi:10.5494/wjh.v2.i4.34
  108. 108. Tarafdar A, Pula G. The Role of NADPH Oxidases and Oxidative Stress in Neurodegenerative Disorders. Int J Mol Sci. 2018;19(12):3824. Published 2018 Nov 30. doi:10.3390/ijms19123824
  109. 109. Schiavone, S., Neri, M., Mhillaj, E. et al. The NADPH oxidase NOX2 as a novel biomarker for suicidality: evidence from human post mortem brain samples. Transl Psychiatry 6, e813 (2016). https://doi.org/10.1038/tp.2016.76
  110. 110. Wang X, Pinto-Duarte A, Sejnowski TJ, Behrens MM. How Nox2-containing NADPH oxidase affects cortical circuits in the NMDA receptor antagonist model of schizophrenia. Antioxid Redox Signal. 2013 Apr 20;18(12):1444-62. doi: 10.1089/ars.2012.4907. Epub 2012 Oct 18. PMID: 22938164; PMCID: PMC3603498.
  111. 111. Stanton RC. Glucose-6-phosphate dehydrogenase, NADPH, and cell survival. IUBMB Life. 2012;64(5):362-369. doi:10.1002/iub.1017
  112. 112. Rajamohan SB, Raghuraman G, Prabhakar NR, Kumar GK. NADPH oxidase-derived H(2)O(2) contributes to angiotensin II-induced aldosterone synthesis in human and rat adrenal cortical cells. Antioxid Redox Signal. 2012;17(3):445-459. doi:10.1089/ars.2011.4176
  113. 113. Buinitskaya Y, Gurinovich R, Clifford G. Wlodaver CG, Kastsiuchenka S. Centrality of G6PD in COVID-19: The Biochemical Rationale and Clinical Implications. Front. Med., 22 October 2020 | https://doi.org/10.3389/fmed.2020.584112
  114. 114. Jain SK, Parsanathan R, Levine SN, Bocchini JA, Holick MF, Vanchiere JA. The potential link between inherited G6PD deficiency, oxidative stress, and vitamin D deficiency and the racial inequities in mortality associated with COVID-19. Free Radic Biol Med. 2020;161:84-91. doi:10.1016/j.freeradbiomed.2020.10.002
  115. 115. Youssef JG, Zahiruddin F, Youssef G, et al. G6PD deficiency and severity of COVID19 pneumonia and acute respiratory distress syndrome: tip of the iceberg?. Ann Hematol. 2021;100(3):667-673. doi:10.1007/s00277-021-04395-1
  116. 116. Guillin OM, Vindry C, Ohlmann T, Chavatte L. Selenium, Selenoproteins and Viral Infection. Nutrients. 2019;11(9):2101. Published 2019 Sep 4. doi:10.3390/nu11092101
  117. 117. Moossavi S, Besharat S, Sharafkhah M, et al. Inverse Association of Plasma Level of Glutathione Peroxidase with Liver Fibrosis in Chronic Hepatitis B: Potential Role of Iron. Middle East Journal of Digestive Diseases. 2016 Apr;8(2):122-130. DOI: 10.15171/mejdd.2016.17.
  118. 118. Rodrigo, R., González, J. & Paoletto, F. The role of oxidative stress in the pathophysiology of hypertension. Hypertens Res 34, 431-440 (2011). https://doi.org/10.1038/hr.2010.264
  119. 119. Hecker PA, Leopold JA, Gupte SA, Recchia FA, Stanley WC. Impact of glucose-6-phosphate dehydrogenase deficiency on the pathophysiology of cardiovascular disease. Am J Physiol Heart Circ Physiol. 2013;304(4):H491-H500. doi:10.1152/ajpheart.00721.2012
  120. 120. Conrad M, Kagan VE, Bayir H, et al. Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev. 2018;32(9-10):602-619. doi:10.1101/gad.314674.118
  121. 121. Li, J., Cao, F., Yin, Hl. et al. Ferroptosis: past, present and future. Cell Death Dis 11, 88 (2020). https://doi.org/10.1038/s41419-020-2298-2
  122. 122. Kwiatkowski DP. How malaria has affected the human genome and what human genetics can teach us about malaria. Am J Hum Genet. 2005;77(2):171-192. doi:10.1086/432519
  123. 123. Bocchetta A. Psychotic mania in glucose-6-phosphate-dehydrogenase-deficient subjects. Ann Gen Hosp Psychiatry. 2003;2(1):6. Published 2003 Jun 13. doi:10.1186/1475-2832-2-6
  124. 124. Manjurano A, Sepulveda N, Nadjm B, et al. African glucose-6-phosphate dehydrogenase alleles associated with protection from severe malaria in heterozygous females in Tanzania. PLoS Genet. 2015;11(2):e1004960. Published 2015 Feb 11. doi:10.1371/journal.pgen.1004960
  125. 125. Fibach E, Dana M. Oxidative Stress in β-Thalassemia. Mol Diagn Ther. 2019 Apr;23(2):245-261. doi: 10.1007/s40291-018-0373-5. PMID: 30484264.
  126. 126. Chirico EN, Pialoux V. Role of oxidative stress in the pathogenesis of sickle cell disease. IUBMB Life. 2012 Jan;64(1):72-80. doi: 10.1002/iub.584. Epub 2011 Nov 30. PMID: 22131167.
  127. 127. Weiss MG. The interrelationship of tropical disease and mental disorder: conceptual framework and literature review (Part I--Malaria). Cult Med Psychiatry. 1985 Jun;9(2):121-200. doi: 10.1007/BF00117368. PMID: 4017618.
  128. 128. Paisseau G. Malaria during the War. Lancet (1919) pp.749-751 pp.
  129. 129. Singh VB, Kumar H, Meena BL, Chandra S, Agrawal J, Kanogiya N. Neuropsychiatric Profile in Malaria: An Overview. J Clin Diagn Res. 2016;10(7):OC24-OC28. doi:10.7860/JCDR/2016/19035.8169
  130. 130. Kim HK, Andreazza AC, Elmi N, Chen W, Young LT. Nod-like receptor pyrin containing 3 (NLRP3) in the post-mortem frontal cortex from patients with bipolar disorder: A potential mediator between mitochondria and immune-activation. J Psychiatr Res. 2016 Jan;72:43-50. doi: 10.1016/j.jpsychires.2015.10.015. Epub 2015 Oct 26. PMID: 26540403.
  131. 131. Wang X, Pinto-Duarte A, Sejnowski TJ, Behrens MM. How Nox2-containing NADPH oxidase affects cortical circuits in the NMDA receptor antagonist model of schizophrenia. Antioxid Redox Signal. 2013 Apr 20;18(12):1444-62. doi: 10.1089/ars.2012.4907. Epub 2012 Oct 18. PMID: 22938164; PMCID: PMC3603498.
  132. 132. Giridharan VV, Scaini G, Colpo GD, et al. Clozapine Prevents Poly (I:C) Induced Inflammation by Modulating NLRP3 Pathway in Microglial Cells. Cells. 2020;9(3):577. Published 2020 Feb 28. doi:10.3390/cells9030577
  133. 133. Siu KL, Yuen KS, Castaño-Rodriguez C, Ye ZW, Yeung ML, Fung SY, Yuan S, Chan CP, Yuen KY, Enjuanes L, Jin DY. Severe acute respiratory syndrome coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J. 2019 Aug;33(8):8865-8877. doi: 10.1096/fj.201802418R. Epub 2019 Apr 29. PMID: 31034780; PMCID: PMC6662968.
  134. 134. Shah A. Novel Coronavirus-Induced NLRP3 Inflammasome Activation: A Potential Drug Target in the Treatment of COVID-19. Front Immunol. 2020;11:1021. Published 2020 May 19. doi:10.3389/fimmu.2020.01021
  135. 135. Argañaraz GA, Palmeira JDF, Argañaraz ER. Phosphatidylserine inside out: a possible underlying mechanism in the inflammation and coagulation abnormalities of COVID-19. Cell Commun Signal. 2020 Dec 27;18(1):190. doi: 10.1186/s12964-020-00687-7. PMID: 33357215; PMCID: PMC7765775.
  136. 136. Hylén U, Eklund D, Humble M, Bartoszek J, Särndahl E, Bejerot S. Increased inflammasome activity in markedly ill psychiatric patients: An explorative study. J Neuroimmunol. 2020 Feb 15;339:577119. doi: 10.1016/j.jneuroim.2019.577119. Epub 2019 Nov 26. PMID: 31786499.
  137. 137. Eda S, Sherman IW. Cytoadherence of malaria-infected red blood cells involves exposure of phosphatidylserine. Cell Physiol Biochem. 2002;12(5-6):373-384. doi: 10.1159/000067908. PMID: 12438774.
  138. 138. Koka S, Lang C, Boini KM, Bobbala D, Huber SM, Lang F. Influence of chlorpromazine on eryptosis, parasitemia and survival of Plasmodium berghe infected mice. Cell Physiol Biochem. 2008;22(1-4):261-268. doi: 10.1159/000149804. Epub 2008 Jul 25. PMID: 18769053.
  139. 139. Delvecchio R, Higa LM, Pezzuto P, et al. Chloroquine, an Endocytosis Blocking Agent, Inhibits Zika Virus Infection in Different Cell Models. Viruses. 2016;8(12):322. Published 2016 Nov 29. doi:10.3390/v8120322
  140. 140. Wang J, Chen S, Bihl J. Exosome-Mediated Transfer of ACE2 (Angiotensin-Converting Enzyme 2) from Endothelial Progenitor Cells Promotes Survival and Function of Endothelial Cell. Oxid Med Cell Longev. 2020 Jan 18;2020:4213541. doi: 10.1155/2020/4213541. PMID: 32051731; PMCID: PMC6995312.
  141. 141. Viskin S, Berger M, Ish-Shalom M, Malov N, Tamari M, Golovner M, Kehati M, Zeltser D, Roth A. Intravenous chlorpromazine for the emergency treatment of uncontrolled symptomatic hypertension in the pre-hospital setting: data from 500 consecutive cases. Isr Med Assoc J. 2005 Dec;7(12):812-815. PMID: 16382707.
  142. 142. Chinevere TD, Murray CK, Grant E Jr, Johnson GA, Duelm F, Hospenthal DR. Prevalence of glucose-6-phosphate dehydrogenase deficiency in U.S. Army personnel. Mil Med. 2006 Sep;171(9):905-907. doi: 10.7205/milmed.171.9.905. PMID: 17036616.
  143. 143. Doumas M, Patoulias D, Katsimardou A, Stavropoulos K, Imprialos K, Karagiannis A. COVID19 and increased mortality in African Americans: socioeconomic differences or does the renin angiotensin system also contribute? [published online ahead of print, 2020 Jul 15]. J Hum Hypertens. 2020;1-4. doi:10.1038/s41371-020-0380-y
  144. 144. Mata-Greenwood E, Chen DB. Racial differences in nitric oxide-dependent vasorelaxation. Reprod Sci. 2008;15(1):9-25. doi:10.1177/1933719107312160
  145. 145. Lavender, N.A., Benford, M.L., VanCleave, T.T. et al. Examination of polymorphic glutathione S-transferase (GST) genes, tobacco smoking and prostate cancer risk among Men of African Descent: A case-control study. BMC Cancer 9, 397 (2009). https://doi.org/10.1186/1471-2407-9-397
  146. 146. Hutchings A, Purcell WM, Benfield MR. Peripheral blood antigen-presenting cells from African-Americans exhibit increased CD80 and CD86 expression. Clin Exp Immunol. 1999;118(2):247-252. doi:10.1046/j.1365-2249.1999.01051.x
  147. 147. Akaberi D, Krambrich J, Ling J, et al. Mitigation of the replication of SARS-CoV-2 by nitric oxide in vitro [published online ahead of print, 2020 Sep 21]. Redox Biol. 2020;37:101734. doi:10.1016/j.redox.2020.101734
  148. 148. Xiao J, Cohen P, Stern MC, Odedina F, Carpten J, Reams R. Mitochondrial biology and prostate cancer ethnic disparity. Carcinogenesis. 2018 Dec 13;39(11):1311-1319. doi: 10.1093/carcin/bgy133. PMID: 30304372; PMCID: PMC6292412.
  149. 149. Feairheller DL, Park JY, Sturgeon KM, et al. Racial differences in oxidative stress and inflammation: in vitro and in vivo. Clin Transl Sci. 2011;4(1):32-37. doi:10.1111/j.1752-8062.2011.00264.x
  150. 150. Miripour ZS, Sarrami-Forooshani R, Sanati H, et al. Real-time diagnosis of reactive oxygen species (ROS) in fresh sputum by electrochemical tracing; correlation between COVID-19 and viral-induced ROS in lung/respiratory epithelium during this pandemic. Biosens Bioelectron. 2020;165:112435. doi:10.1016/j.bios.2020.112435
  151. 151. Ibrahim H, Perl A, Smith D, et al. Therapeutic blockade of inflammation in severe COVID-19 infection with intravenous N-acetylcysteine. Clin Immunol. 2020;219:108544. doi:10.1016/j.clim.2020.108544
  152. 152. de Queiroz TM, Monteiro MM, Braga VA. Angiotensin-II-derived reactive oxygen species on baroreflex sensitivity during hypertension: new perspectives. Front Physiol. 2013;4:105. Published 2013 May 13. doi:10.3389/fphys.2013.00105
  153. 153. Guimarães, D. D., Carvalho, C. C., and Braga, V. A. (2012). Scavenging of NADPH oxidase-derived superoxide anions improves depressed baroreflex sensitivity in spontaneously hypertensive rats. Clin. Exp. Pharmacol. Physiol. 39, 373-378.
  154. 154. Braga, V. A. (2010). Dietary salt enhances angiotensin-II-induced superoxide formation in the rostral ventrolateral medulla. Auton. Neurosci. 155, 14-18
  155. 155. Inukai T, Yoshida N, Wakabayashi S, Inukai Y, Matsutomo R, Takanashi K, Nakamachi T, Takebayashi K, Aso Y, Takahashi K. Angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers effectively and directly potentiate superoxide scavenging by polymorphonuclear leukocytes from patients with type 2 diabetes mellitus. Am J Med Sci. 2005 May;329
  156. 156. Cianchetti S, Del Fiorentino A, Colognato R, Di Stefano R, Franzoni F, Pedrinelli R. Anti-inflammatory and anti-oxidant properties of telmisartan in cultured human umbilical vein endothelial cells. Atherosclerosis. 2008 May;198(1):22-28. doi: 10.1016/j.atherosclerosis.2007.09.013. Epub 2007 Oct 22. PMID: 17950296.
  157. 157. Heringer-Walther S, Batista EN, Walther T, Khosla MC, Santos RA, Campagnole-Santos MJ. Baroreflex improvement in shr after ace inhibition involves angiotensin-(1-7). Hypertension. 2001 May;37(5):1309-1314. doi: 10.1161/01.hyp.37.5.1309. PMID: 11358946
  158. 158. Eguchi K, Shimizu M, Hoshide S, Shimada K, Kario K. A bedtime dose of ARB was better than a morning dose in improving baroreflex sensitivity and urinary albumin excretion--the J-TOP study. Clin Exp Hypertens. 2012;34(7):488-492. doi: 10.3109/10641963.2012.666604. Epub 2012 Apr 25. PMID: 22533496.
  159. 159. Williams SF, Nicholas SB, Vaziri ND, Norris KC. African Americans, hypertension and the renin angiotensin system. World J Cardiol. 2014;6(9):878-889. doi:10.4330/wjc.v6.i9.878
  160. 160. Flack JM, Mensah GA, Ferrario CM. Using angiotensin converting enzyme inhibitors in African-American hypertensives: a new approach to treating hypertension and preventing target-organ damage. Curr Med Res Opin. 2000;16(2):66-79. PMID: 10893650.

Written By

Adonis Sfera, Carolina Osorio, Jose E. Campo Maldonado, Afzaal Jafri, Aaron D. Chokka, Carlos Manuel Zapata Martín del Campo and Zisis Kozlakidis

Submitted: 25 November 2020 Reviewed: 01 March 2021 Published: 03 May 2021