Next Article in Journal
Flavonol Composition and Antioxidant Activity of Onions (Allium cepa L.) Based on the Development of New Analytical Ultrasound-Assisted Extraction Methods
Next Article in Special Issue
Effect of Antioxidant Therapy on Oxidative Stress in Vivo
Previous Article in Journal
FK866 Protects Human Dental Pulp Cells against Oxidative Stress-Induced Cellular Senescence
Previous Article in Special Issue
Multiple Effects of Ascorbic Acid against Chronic Diseases: Updated Evidence from Preclinical and Clinical Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Role of Antioxidant and Anti-Inflammatory Therapies to Prevent Severe SARS-Cov-2 Complications

by
Anna M. Fratta Pasini
*,
Chiara Stranieri
,
Luciano Cominacini
and
Chiara Mozzini
Section of General Medicine and Atherothrombotic and Degenerative Diseases, Department of Medicine, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
*
Author to whom correspondence should be addressed.
Submission received: 31 December 2020 / Revised: 3 February 2021 / Accepted: 4 February 2021 / Published: 10 February 2021
(This article belongs to the Special Issue Effect of Antioxidant Therapy on Oxidative Stress In Vivo)

Abstract

:
The coronavirus disease 2019 (COVID-19) pandemic is caused by a novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2). Here, we review the molecular pathogenesis of SARS-CoV-2 and its relationship with oxidative stress (OS) and inflammation. Furthermore, we analyze the potential role of antioxidant and anti-inflammatory therapies to prevent severe complications. OS has a potential key role in the COVID-19 pathogenesis by triggering the NOD-like receptor family pyrin domain containing 3 inflammasome and nuclear factor-kB (NF-kB). While exposure to many pro-oxidants usually induces nuclear factor erythroid 2 p45-related factor2 (NRF2) activation and upregulation of antioxidant related elements expression, respiratory viral infections often inhibit NRF2 and/or activate NF-kB pathways, resulting in inflammation and oxidative injury. Hence, the use of radical scavengers like N-acetylcysteine and vitamin C, as well as of steroids and inflammasome inhibitors, has been proposed. The NRF2 pathway has been shown to be suppressed in severe SARS-CoV-2 patients. Pharmacological NRF2 inducers have been reported to inhibit SARS-CoV-2 replication, the inflammatory response, and transmembrane protease serine 2 activation, which for the entry of SARS-CoV-2 into the host cells through the angiotensin converting enzyme 2 receptor. Thus, NRF2 activation may represent a potential path out of the woods in COVID-19 pandemic.

1. Introduction

The coronavirus disease 2019 (COVID-19) pandemic is caused by a novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) [1].
SARS-CoV-2 is an enveloped, non-segmented, positive sense RNA virus, widely distributed in humans and other mammals [2,3]. SARS-CoV-2 is dissimilar from the coronaviruses recognized to induce the ordinary cold, but it has been shown to have the same characteristics as the zoonotic SARS coronavirus (SARS-CoV) [4] and the Middle East respiratory syndrome (MERS) coronavirus [5]. Patients affected by COVID-19 often display no symptoms or mild symptoms (fever, cough, myalgia, and fatigue) and usually have a good prognosis. Many of these cases, however, progress to a more severe form of the illness, especially in older men experiencing other contemporary serious diseases [2,6,7,8]. Severe patients can suffer from symptoms correlated with lung [2,8,9], heart [8,10,11], kidney [8,12,13], neurological [14,15], gastrointestinal [16] and liver [9,16,17,18] injuries. Furthermore, there may be immune [9,12,19,20] and coagulation [21,22] impairment. Globally, as of December 27, 2020, there have been 79,232,555 confirmed COVID-19 cases, including 1,754,493 deaths [23].
Angiotensin converting enzyme 2 (ACE2) offers an access receptor for SARS-CoV-2 and SARS-CoV in humans by binding to the viral membrane spike (S) protein [24,25]. The quick recognition of ACE2 as SARS-CoV-2 receptor is mostly attributable to its recognition as the receptor for SARS-CoV about 17 years ago. In that case, ACE2 was recognized as the functional receptor for SARS-CoV after the fusion protein gene of SARS-CoV was reported [26]. By means of in vitro studies, Li et al. [27] found that: (1) ACE2 attached to the SARS-CoV S1 protein; (2) a soluble variety of ACE2, but not ACE1, inhibited the binding of the S1 protein with ACE2; (3) SARS-CoV reproduced in a very intense manner in ACE2-transfected, but not mock-transfected, cells. Furthermore, studies in vivo have clearly shown that ACE2 is a pivotal SARS-CoV receptor [28]. Here, we review the molecular pathogenesis of SARS-CoV-2 and its relationship with oxidative stress (OS) and inflammation. Furthermore, we analyze the potential role of antioxidant and anti-inflammatory therapies to prevent severe complications.

2. SARS-CoV-2 Cell Entry Mechanisms

2.1. SARS-CoV-2 Structural Basis

Like SARS-CoV, SARS-CoV-2 has four principal structural proteins: spike (S), envelope (E), membrane (M) and nucleocapsid (N), together with several additional proteins [29,30] (Figure 1). The S glycoprotein is a transmembrane protein (molecular weight of about 150 kDa) found in the virus outer portion [31]. Like SARS-CoV, S protein occurs as a trimer, with three receptor-binding S1 heads being placed on top of a membrane fusion S2 stalk [31] (Figure 1). S1, which binds to the peptidase domain of ACE2, is called the receptor-binding domain (RBD), while S2 catalyzes the membrane fusion, thus releasing the genetic material into the cells [31]. The crystal structures of the RBD of the S protein of SARS-CoV-2, both non-complexed [32] (protein data bank code 6VXX, https://www.rcsb.org (accessed on 31 December 2020)) or complexed with human ACE2 [33] (protein data bank code 6M0J, https://www.rcsb.org (accessed on 31 December 2020)) have been published previously. Recent studies, however, have established that there are slight differences between SARS-CoV-2 and SARS-CoV in receptor recognition [34]; these dissimilarities allow SARS-CoV-2 RBD to possess a slightly higher ACE2 receptor affinity than RBD of SARS-CoV [31], even though it results in being less accessible [32,35]. To retain its elevated infectivity despite a low accessibility, SARS-CoV-2 uses activation of host proteases, and this process crucially determines the infectivity and pathogenesis of SARS-CoV-2 infection [31]. In this context, it has previously been established that the pre-activation of furin, a host proprotein convertase [35,36], increases SARS-CoV-2 entrance into cells expressing ACE2 receptor by binding to a polybasic sequence motif at the S1/S2 border of the virus [31]. Furin-cleaved substrates then link to neuropilin-1 (NRP1), facilitating SARS-CoV-2 infectivity [36,37]. Moreover, transmembrane protease serine 2 (TMPRSS2) and lysosomal cathepsins, in addition to forcing SARS-CoV-2 entrance, have an additional impact with furin on SARS-CoV-2 entry [31]. Entered-SARS-CoV-2 will subsequently release its genomic material in the cytoplasm and be translated into the nuclei [38].

2.2. Structural Basis and Function of ACE2 Receptor

The renin-angiotensin system (RAS) plays a role in controlling blood volume and systemic vascular resistance, which at the same time affect cardiac output and arterial pressure [39]. ACE, a dipeptidyl carboxypeptidase in the RAS, converts the inactive angiotensin (Ang) I into the active and effective vasoconstrictor Ang II and inhibits the vasodilator Bradykinin [40]. ACE2 counterbalances ACE to a great extent by converting Ang I into Ang 1–9, an inert variety of Ang. It can also break down and hydrolyze the vasoconstrictor Ang II, into Ang 1–7, which acts as a strong vasodilator [41]. The ACE2 crystal structure and RBD of the S protein of SARS-CoV-2 complexed with human ACE2 have previously been reported [33,42] (protein data bank codes 1R42 and 6M0J, respectively, https://www.rcsb.org (accessed on 31 December 2020)). As just reviewed [43,44], ACE2 has multiple crucial protecting roles against hypertension, cardiovascular and lung diseases, and diabetes mellitus. Furthermore, the control of gut dysbiosis and vascular permeability by ACE2 has come out as an intrinsic mechanism of pulmonary hypertension and diabetes-related cardiovascular complications [44].
Very recently, ACE2 has been garnering widespread interest as a functional SARS-CoV-2 and SARS-CoV virus receptor by binding to the viral S protein, in this way contributing to pathogenesis of SARS [11,24,25,27]. ACE2 is ubiquitously expressed, with the highest levels in the epithelial cells of the lung, kidney and cardiomyocytes [45], although there is no lack of discordant voices, mostly for lung tissue [46]. Furthermore, recent studies based on single-cell RNA-sequence (scRNA-seq) data analysis have reported that ACE2 is widespread in many organs, including the lungs, heart, esophagus, kidneys, bladder, ileum, oral mucosa, and, particularly in the case of type II alveolar cells, cardiomyocytes, kidney proximal tubule cells, ileum and esophagus epithelial cells, and bladder urothelial cells [47]. Thank to this diffuse presence, ACE2 is involved in virus infection and diffusion. In addition, it has previously been found that infection with SARS-CoV and SARS-CoV-2 causes ACE2 shedding with subsequent downregulation of surface ACE2 expression [28,48]. In this context, in a small group of severe COVID-19 patients, Ang II plasma concentration was found to be significantly higher than in healthy controls [49], strengthening the hypothesis of a direct link between tissue ACE2 downregulation with systemic RAS imbalance.
As shown in Figure 2, recent evidence has shown that ectodomain shedding of ACE2 is mediated by ADAM17 (a disintegrin and metalloproteinase17), which in turn is upregulated by endocytosed SARS-CoV-2 S proteins [50] and other mechanisms [51,52,53,54].
The available body of facts indicates that Ang II binding to AT1R also controls the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [NOX] [54,55], one of the most important determinants of reactive oxygen species (ROS) generation. Hence, the SARS-CoV-2-induced ACE2 downregulation increases the binding of Ang II to AT1R, which, by triggering NOX, causes oxidative stress (OS) and inflammation in accordance with the COVID-19 severity [46].

3. TMPRSS2 and SARS-CoV-2

One crucial discovery in learning how SARS-CoV-2 enters into the cells involves the role of TMPRSS2, a cell-surface protein [56] (Figure 2) that was identified in 2001 in the epithelia of the gastrointestinal, urogenital, and respiratory tracts of mouse and humans, although TMPRSS2 expression in human dominates in the prostate [57]. The crystal structure of TMPRSS2 has previously been published ([58], protein data bank code 5AFW, https://www.rcsb.org (accessed on 31 December 2020)). As for the expression of TMPRSS2 in the lung and bronchial branches, a very recent study using the scRNA-seq method showed that the highest expression of TMPRSS2 was in alveolar type 2 cells. Interestingly, these cells also presented the greatest expression of ACE2 [59,60,61,62]. Since SARS-CoV-2 has a furin cleavage site in its S protein, with the potential to increase SARS-CoV-2 binding to ACE2 receptor [31], the authors also detected a preference for co-expression for any association of ACE2, TMPRSS2 and/or furin expression [59]. Accordingly, the priming of SARS-CoV-2 S protein by furin would hypothetically make many more cells susceptible to infection, as compared to S protein priming by TMPRSS2 alone [59]. Furin activity first causes the generation of two non-covalently associated proteins, S1 and S2 [32,60,61], with the TMPRSS2 further priming S2 [57]. Then, the C terminus of the S1 protein may bind to the NRP1, which significantly potentiates SARS-CoV-2 infectivity [36,37]. In this context, however, it has been reported that cells expressing NRP1 alone only play a small part in SARS-CoV-2 infection [36], whereas its co-expression with ACE2 and TMPRSS2 greatly intensified infection [36].
Finally, it has to be pointed out that TMPRSS2 has been identified in prostate cancer, and that its expression was upregulated by androgens [62,63]. Previous reports showed that androgen receptors are expressed in the human respiratory tract epithelium, mainly in type 2 alveolar and bronchial epithelial cells [64]. Since growing data support the concept that male gender is a factor associated with a significantly increased risk of severe events and death from COVID-19 [12,65,66], the strong up-regulation of TMPRSS2 by androgens raises the theory that the male prevalence in the COVID-19 pandemic could partially be explained by androgen-driven TMPRSS2 increase [67]. The available data on this point, however, are discrepant [67], and further studies are needed to fully clarify this topic.

4. Oxidative Stress and Inflammation Associated with SARS-CoV-2 Infection

4.1. Oxidative Stress (OS) in SARS-CoV-2 Infection

It is known that OS arises whenever there is an imbalance between ROS formation and antioxidant defenses. Alterations of the redox state towards oxidant conditions in infected cells is one of the key events in respiratory viral infections that is linked to inflammation and subsequent tissue damage [68,69,70]. Recent evidence indicates that OS play a crucial role also in COVID-19 infection [71,72,73,74,75]. Several in vitro and in vivo studies have shown that ROS overproduction induced by respiratory viruses is partially mediated by the activity of NOX (reviewed in [69]). As reported above, ACE2 shedding caused by SARS-CoV-2 fusion may be strictly related to RAS imbalance [43,47], and there is now evidence that Ang II controls NOX activation [54,55] (Figure 3). It has been suggested that NOX2 is a key event in killing bacteria and fungi, but it does not efficiently function against viruses [71]. In this regard, a recent study shows that OS induced by NOX2 activation is linked with severe clinical outcome and thrombotic events in COVID-19 patients [76]. ACE2 downregulation and OS are also associated with endothelial dysfunction via NOX activation and reduced availability of nitric oxide [77]. Furthermore, oxidized phospholipids (OxPLs), which are a product of OS and have been detected in the lungs of SARS-CoV patients [78], were found to be one of the main triggers of acute lung injury. As a matter of fact, OxPLs were shown to promote tissue factor expression [78], to activate endothelial cells to recruit monocytes [79,80], and to trigger macrophage activation through Nuclear Factor-κB (NF-kB) pathway [78]. It remains to be elucidated whether analogous pathways are also involved in SARS CoV-2 infection.
It is well recognized that the levels of cellular free iron must be tightly regulated to avoid ROS generation via the Fenton reaction [81]. Upon SARS-CoV-2 infection, IL-6 in the cytokine storm increases ferritin and the production of hepcidin, which plays a main role in iron regulation. Since iron is sequestered by hepcidin in the enterocytes and macrophages, intracellular ferritin is augmented, leading to a reduced iron efflux from the cells. The stored iron may increase intracellular labile iron (II) pool and Fenton reaction, producing lipid ROS, and lead to ferroptosis, a novel form of regulated cell death [81]. In COVID-19 patients, the documented iron metabolism alterations may cause iron accumulation and overload, triggering ferroptosis in the cells of multiple organs [82,83].
Many lines of evidence show that viruses may also generate OS per se [69,70]. With regard to SARS-CoV, the viral protease 3CLpro has been previously shown to increase ROS generation in HL-CZ cells, with subsequent cell apoptosis and NF-kB-activation [84]. Another SARS-CoV protease, the 3a protein, has been linked with mitochondrial cell death pathway activation by triggering OS [69].
The mitochondrial respiratory chain is the main and most significant source of cellular ROS. However, while mitochondrial ROS production was once seen as merely an accidental by-product of oxygen metabolism of mitochondrial respiratory chain, it is now clear that ROS contribute to various signaling pathways [85]. Depending on the context and triggering stimuli, mitochondrial ROS production can lead to different cellular responses such as adaptation to hypoxia, differentiation, autophagy, inflammation, or to an immune response [86]. In general, viruses can modify mitochondrial dynamics in a highly specific manner so that they can successfully replicate [87]. Among the different mechanisms implicated, there are mitochondrial DNA damage, changes in mitochondrial membrane potential, variations in mitochondrial metabolic pathways and calcium homeostasis, modifications in number and distribution of mitochondria into the cells, weakening of antioxidant defense, and augmented OS [87,88]. Upon infection, viruses completely rely on host cell molecular machinery to survive and replicate [87,88]. Mitochondria defend host cells from SARS-CoV-2 virus through several mechanisms including cellular apoptosis, ROS production, autophagy, mitochondrial antiviral signaling system (MAVS) activation, DNA-dependent immune activation, and other things [89]. Current knowledge of how SARS-CoV-2 infection affects mitochondria and their ROS generation is limited. A prior study on SARS-CoV [90] showed that open reading frame-9b (Orf9b), one of the accessory proteins of the virus [91], alters host cell mitochondria morphology, disrupts MAVS, inhibits interferon (IFN) production and enhances autophagy, a cellular mechanism activated by ROS [92]. Consistent with the findings of SARS-CoV, Gordon et al. [93] recently reported that SARS-CoV-2 Orf9b interacts with mitochondrial translocase of outer membrane (TOM)70, although the functional consequences of this association were not examined. Very recently, Jiang et al. [94] reported that SARS-CoV-2 Orf9b localizes to the membrane of mitochondria and suppresses IFN-I response through association with TOM70. The altered activity of TOM70, by reducing constitutive calcium transfer to mitochondria, dampens mitochondrial respiration, affects cell bioenergetics, and induces autophagy [95].
During viral infections beyond an over-production of ROS, there is a decreased antioxidant defense, mainly Glutathione (GSH) depletion, in the host cells that directly or indirectly favor viral replication [96]. GSH, a tripeptide consisting of cysteine, glycine, and glutamate, is the main intracellular antioxidant that applies an efficient buffering role against ROS, through the thiol group of its cysteine which oxidizes to the disulfide form, then reduced back to the thiol form by glutathione reductase [70].
It has a principal role in cellular signaling and processes, as well as innate immune response to viruses [70].
A significant elevation in blood serum GSH reductase, derived from OS imbalance, was found in COVID-19 patients, especially when admitted to the intensive care unit [97]. Additionally, mounting evidence supports the concept that the reduced levels of GSH may underlie the COVID-19 severe clinical outcome and death [98].

4.2. Cross Talks between Oxidative Stress and Inflammation in SARS-CoV-2 Infection

Several studies have demonstrated that SARS-CoV-2 infection and the destruction of lung cells causes a local immune response, recruiting macrophages and monocytes that reply to the infection, release cytokines and prime adaptive T and B cell immune responses. In most patients, this process overcomes the infection. However, sometimes, a dysfunctional immune response occurs, which leads to a cytokine storm that mediates general lung inflammation [2,99,100]. Increased plasma concentrations of inflammatory markers such as C-reactive protein and ferritin, of many cytokines such as TNF-alpha, IL-1beta, IL-6 and IL-8, and chemokines such as MCP1, together with increased neutrophils/lymphocytes ratio [11,99,100], have been associated with gravity of SARS-CoV-2 infection and death [2,19,101].
SARS-CoV-2 infection in type 2 alveolar and other cells activates NOD-like receptor protein 3 (NLRP3), an element of the innate immune system that acts as a pattern recognition receptor that recognizes damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) [102] and takes part in multiprotein complexes called inflammasomes, which bring together sensor proteins (like NLRP3) [103,104]. NLRP3 inflammasome is very often associated with cellular death by apoptosis and pyroptosis [105,106,107], an inflammatory form of programmed cell death [108] that releases large amounts of pro-inflammatory mediators [109]. Accumulating data have established a causal role between the pyroptosis of alveolar type 2, endothelial and immune cells and the progression of lung damage [110,111,112,113,114,115]. The contemporary activation of alveolar macrophages further produces large amounts of proinflammatory cytokines and chemokines [116,117], which activate endothelial cells [118,119], platelets [120,121] and neutrophils [122,123] generating platelet neutrophil complexes at endothelium surface [124,125]. This sequestration of platelet neutrophil complexes in the pulmonary vasculature is the prelude of a highly inflammatory and pro-coagulant situation, a state called immunothrombosis [119,126,127]. Convincing evidence shows that immunothrombosis is a pivotal determinant of micro-thrombi and micro-emboli generation in the alveolar capillary circulation [128,129], of fibrin deposition within the alveoli, and in some cases of disseminated intravascular coagulation [130,131,132]. Furthermore, the huge associated increase of activated neutrophils in lung interstitial tissue and alveoli [133] can discharge high levels of extremely cytotoxic neutrophil extracellular traps [133]. These events play a crucial part in determining intra-lung cytokine storm and the consequent tissue damage that is a peculiarity of ARDS, an inflammatory disease with pulmonary epithelial and capillary endothelial cells dysfunction, alveolar macrophages and neutrophils infiltration, cell death, and fibrosis [134].
While it is likely that lung and other tissue damages in SARS-CoV-2 infection are the results of multifactorial mechanisms, very recent studies indicate that ROS may play a major role in the initiation and progression of this inflammatory process [135,136]. In this context, it has been reported that OS triggers the NLRP3 inflammasome [137,138]. Although it is conceivable that other pathological pathways participate in NLRP3 induction [139,140,141], OS activates NLRP3 inflammasome through NF-kB and thioredoxin interacting/inhibiting protein [135,136,137,138,142,143] activation. In addition, NF-kB up-regulates IL-18 and IL-1beta expression, further increasing NLRP3 inflammasome [141,143,144], as shown in Figure 3. This OS-induced overactivation of NLRP3 inflammasome may play a key role in the pathogenesis of severe SARS-CoV-2 infection. In fact, when the innate response cannot clear the infection, the resulting NLRP3 hyperactivation is harmful, leading to perturbation of mitochondrial function, the release of DAMPS and mounting pyroptosis [102,103,104] determining virus propagation and massive destruction of affected tissues [145,146].

5. Rationale for Antioxidant and Anti-Inflammatory Therapies against COVID-19 Complications

5.1. Radical Scavengers

Modulation of the intracellular redox state is a pivotal strategy that viruses use to manipulate host cell machinery to their advantage [68]. Accordingly, recent studies have focused on redox-sensitive pathways as novel cell-based targets for therapies designed to stop both viral replication and virus-induced inflammation. Since respiratory viruses not only improve ROS production but also impair cellular defense systems, the use of radical scavengers has long been considered to be a potential therapeutical approach [69,70].
In the COVID-19 pandemic, the search for alternative therapies for the treatment of coronavirus diseases is of great importance; in this context, antioxidant therapies have been proposed as a potential treatment, preventive and/or adjuvant against SARS-CoV-2.
The most encouraging compounds comprise GSH and its precursor N-acetylcysteine (NAC). NAC is a natural antioxidant derived from plants especially from the Allium species, whose thiol group directly scavenges ROS and helps GSH synthesis [147]. Since NAC is applied in a broad range of conditions to restore GSH depletion it has been suggested as a nutraceutical that might aid the control of RNA viruses including influenza and coronavirus [148].
It is well recognized that the interaction of viral S protein with ACE2 is an important step in the viral replication cycle [24,25]. The RBD of the viral S protein and ACE2 have several cysteine residues [149,150]; interestingly, it has recently been found that the binding affinity is significantly impaired when all the disulfide bonds of both ACE2 and SARS-CoV/CoV-2 S proteins are reduced to thiol groups [149,150]. These facts are consistent with the view that the reduction of disulfides into sulfhydryl groups completely impairs the binding of SARS-CoV/CoV-2 S protein to ACE2 and provide a molecular basis for the COVID-19 infection severity due to OS [149,150].
Based on the protective role of NAC in experimental models of influenza and other viruses [151,152], it has recently been suggested that NAC may be used both in the COVID-19 prevention and in therapy [153]. Recently, NAC has been demonstrated to also exert protective mechanisms against a variety of COVID-19 associated conditions including cardiovascular diseases [154]. Administration of NAC has also been considered among the possible strategies aimed at protecting endothelial function and restricting microthrombosis in severe forms of the COVID-19 disease [155]. A potential role of NAC and copper in combination with candidate antiviral treatments against SARS-CoV-2, such as remdesivir, has been hypothesized based on a systematic literature search [156]. Clearly, these possible anti-COVID-19 mechanisms and properties of NAC need to be confirmed in controlled clinical trials [157,158]. In particular, the results of “A Study of N-acetylcysteine in Patients with COVID-19 Infection” (NCT04374461) aimed at evaluating the effect of NAC (iv; 6 g/day) administration as an adjuvant treatment in patients with severe COVID-19 symptoms will help to corroborate the potential therapeutic properties of this thiol in COVID 19 patients. The patients were enrolled into two separate arms and the mechanically ventilated and/or managed in a critical-care arm is closed to accrual as of September 2020 [159].
A further mechanism that has recently been proposed is the possibility that NAC further improves the stimulation of Nuclear factor erythroid 2 p45-related factor2 (NRF2) by OS, which promotes the transcription of phase II enzyme genes and downregulates inflammation [160]. At the same time, NAC prevents the OS-mediated activation of NF-κB and biochemical pathways upregulating pro-inflammatory genes [161]. NAC also reduced the intracellular hydrogen peroxide concentration and restored the intracellular total thiol contents by impeding NF-κB translocation to the cellular nucleus and phosphorylation of p38 mitogen-activated protein kinase [162].
Taken together, the results of experimental and clinical studies available so far indicate that NAC acts in a variety of potential therapeutic target pathways involved in the pathophysiology of SARS-CoV-2 infection.
It is well recognized that during viral infections, intracellular GSH depletion is a common event that is central for viral replication [97], and several in vitro and in vivo studies have found that GSH administration blocks viral replication through redox state modulation [70]. An improving GSH molecule is I-152, a combination of NAC and s-acetyl-mercaptoethylamine (cysteamine, MEA) that can release NAC and MEA thus increasing GSH. Its antiviral efficacy has been evidenced in in vitro and in vivo models [163]. Interestingly, a case report study showed that the repetitive use of both 2000 mg of oral administration and intravenous injection of GSH was effective at relieving COVID-19 severe respiratory symptoms, demonstrating for the first time the usefulness of this antioxidant therapy for COVID-19 patients [164].
As far as Vitamin C is concerned, its important anti-inflammatory, immunomodulating, antioxidant, antithrombotic and antiviral properties are well known [165] as a contributor in cytokine down-regulation and ROS lowering via attenuation of NF-kB activation.
Vitamin C deficiency in gulonolactone Loxidase-knockout mice [166] showed enhanced Neutrophil Extracellular Traps (NETs) in the lungs of septic animals and increased circulating cell-free DNA suggesting that vitamin C is a novel regulator of NETosis, which is a particular cell death [167] implicated in the response to fighting COVID-19 [168].
The pharmacological effects of Vitamin C that could make it a potential option for prevention and treatment of COVID-19 have recently been reviewed [169]. Clinicians using intravenous Vitamin C in severely ill COVID-19 patients have reported positive clinical effects upon administration of 3 g every 6 h, together with steroids and anti-coagulants [170]. There are currently several clinical trials registered on Clinicaltrials.gov investigating Vitamin C (oral or intravenous) with or without other treatments for COVID-19. The largest registered trial is the Lessening Organ Dysfunction with Vitamin C-COVID (LOVIT-COVID) trial in Canada, which is recruiting 800 patients who are randomly assigned to Vitamin C (intravenous, 50 mg/kg every 6 h) or a placebo for 96 h, i.e., equivalent to 15 g/day (NCT04401150). This protocol has also been added as a Vitamin C arm in the Randomized, Embedded, Multifactorial Adaptive Platform Trial for Community-Acquired Pneumonia (REMAP-CAP; NCT02735707). The study design provides further rationale for the use of Vitamin C in COVID-19 patients [171].
A case series of 17 COVID-19 patients who were given 1 g of intravenous Vitamin C every 8 h for 3 days reported decreased mortality, decreased intubation and mechanical ventilation need and a significant decrease in inflammatory markers, including ferritin and D-dimer, and a trend towards decreasing FiO2 requirements [172]. These parameters are under investigation in the “Intravenous Vitamin C Administration in Coronavirus (COVID-19) and Decreased Oxygenation (AVoCaDO), NCT04357782” clinical trial in which subjects administered with intravenous Vitamin C are supposed to be at lower risk of respiratory failure worsening and reduced inflammation markers increase. As of October 13, 2020, recruitment has been completed.
Whether or not Vitamin C supplementation will consistently prevent conversion to the critical phase of COVID-19 has yet to be determined, but given its favorable safety profile and low cost, and the frequency of its deficiency in respiratory infections, it may be worthwhile testing patients’ vitamin C status and treating them.

5.2. NRF2 Activators

NRF2 is a leading transcription factor that targets genes coding for antioxidant proteins and detoxification enzymes [161]. NRF2 regulates the basal and induced expression of an array of antioxidant response element (ARE-dependent genes, such as heme-oxygenase (HO)-1) to regulate the physiological and pathophysiological outcomes of oxidant exposure. Under basal conditions, NRF2-dependent transcription is blocked by its negative regulator, Kelch-like enoyl-CoA hydratase-associated protein1 (Keap-1); when cells are exposed to OS or electrophiles, NRF2 accumulates in the nucleus and drives its target genes expression [160].
Several studies support the concept that viral infections interfere with antioxidative systems, causing an imbalance between oxidative and antioxidative status and subsequent oxidative cell injuries [68,69,70,71]. In particular, while exposure to many pro-oxidants induces NRF2 activation and upregulation of ARE gene expression, respiratory viral infections often inhibit NRF2 pathway and/or activate NF-kB transcription factor, resulting in inflammation and oxidative injury [173]. The activation or inhibition of NRF2 in host cells is dependent on factors such as the stage of infection [174] or the peculiar viral propagation mechanisms by which cell death and release of viruses are caused [175]. A first key demonstration that SARS-CoV-2 virus deprives the host cells of this essential cytoprotective pathway stems from the recent evidence indicating that NRF2 pathway was repressed in lung biopsies of patients affected by SARS-CoV-2 infection [176].
There is a reciprocal crosstalk between NRF2 and NF-κB when innate immune cells are enrolled in inflamed tissues [177,178,179]. In in vitro studies, subsequent to infection with SARS-CoV, NF-κB was reported to switch on in mice lungs and in human macrophages; on the contrary, NF-κB inhibition decreased inflammation and ameliorates survival in SARS-CoV-infected mice [180,181]. Therefore, while NRF2 suppression may be associated with high-grade NF-kB activation and consequently with inflammation, activation of NRF2 by specific drugs may delimit NF-κB activity in patients with SARS-CoV-2 infection.
Increasing evidence supports the concept that pharmacological activation of NRF2 may be a promising adjuvant therapy against SARS-CoV-2 infection [182]. In particular, NRF2 inducers may protect against the excessive inflammatory response in COVID-19 patients through different mechanisms: host cell protection, anti-inflammatory phenotype activation, thus preventing uncontrolled proinflammatory cytokines production, pyroptosis and viral propagation inhibition [182].
NRF2 can be triggered by pharmacological inducers that target Keap1; in fact, a lot of NRF2 inducers, including dimethyl fumarate (DMF), sulforaphane, and bardoxolone methyl, are electrophiles that alter cysteine sensors of Keap1 and disarm its repressor function [182].
An important issue is whether NRF2 activators may reduce SARS-CoV-2 replication. In this context, the NRF2 agonists 4-octyl-itaconate (4-OI) and the clinically approved DMF suppress SARS-CoV-2 replication and the expression of associated inflammatory genes in cultured cells [176]. In the opinion of the authors [176], the fact that 4-OI suppressed to a great degree the IFN antiviral response but maintained the capacity to inhibit viral replication and attenuate the inflammatory response suggests the existence of unrecognized cellular pathways that work independently of IFNs.
Many reports have described numerous antiviral effects for HO-1 against a broad spectrum of viruses. In many cases, the mechanism of action of HO-1 products has been recognized, showing direct effects on virus components or cellular processes that interfere with virus replication [183]. Although there are no data so far for targeting HO-1 on SARS-CoV-2, it has been proposed that inducing HO-1 expression may avoid SARS-CoV-2-induced lung complications by means of its antiviral, anti-inflammatory, antithrombotic and antifibrotic properties [184].
Another important point is whether NRF2 can suppress SARS-CoV-2 access into the host cells, and in this scenario, a key role is carried out by TMPRSS2 [56]. PB125, a strong NRF2 inducer, was able to significantly downregulate ACE2 and TMPRSS2 expression in HEPG2 cells [185]. Intriguingly, it also induced a strong upregulation of the human antiprotease plasminogen activator inhibitor-1 (PAI-1) expression, a potent TMPRSS2 inhibitor [186]. Accordingly, the authors suggest that PB125 treatment might reduce the SARS-CoV-2 capacity to bind to a host cell and to provoke S protein activation [185]. In the same study, PB125 was also shown to markedly downregulate genes encoding cytokines [185], many of which were exactly recognized in the cytokine storm seen in lethal cases of COVID-19 [187]. Moreover, it was previously reported that bromhexine, an FDA-approved ingredient in mucolytic cough suppressants, had the capacity to inhibit TMPRSS2 activity and to reduce prostate cancer enlargement and metastases [188]. At present, the mechanism involved in bromhexine-induced TMPRSS2 activity suppression is unknown. However, ambroxol, a metabolite of bromhexine, which has been approved by the FDA and has been established for decades for the treatment of acute and chronic respiratory diseases [189], has also been found to exert an excellent anti-inflammatory and antioxidant activity and to elicit a remarkable induction of NRF2 associated with a concomitant decrease in NF-kB expression in mice [190]. The bromhexine effectiveness in SARS-CoV-2 infection in a small open-label randomized clinical was recently reported by Ansarin et al. [191]. They found that bromhexine administration was associated with a significant reduction in intensive care unit admissions, intubation and death suggesting that TMPRSS2 suppression may contribute to clinically ameliorate SARS-CoV-2 infection (Figure 4).
Finally, DMF, which is now used as an anti-inflammatory drug in relapsing-remitting Multiple Sclerosis [192], could easily be repurposed and verified in clinical trials as a small molecule inhibitor of SARS-CoV-2 replication and inflammation-induced pathology in COVID19 patients.
Likewise, the wealth of safety and efficacy information for other NRF2 activators, such as sulforaphane and bardoxolone methyl, which are now in advanced clinical trials for other indications, offers a clear means for their testing in COVID-19 randomized clinical trials. If confirmed, this therapeutic strategy could be rapidly mobilized to improve recovery and decrease the need for mechanical ventilation in severe COVID-19 patients, helping to relieve the big strain that is currently being experienced by intensive care units worldwide [182].

5.3. Delivery of Soluble ACE2

SARS-CoV-2 infection causes ACE2 shedding from tissue, thus effectively lowering the ACE2 receptor level in infected cells [28,48]. In this regard, it has been suggested that delivery of recombinant ACE2 protein may be a treatment to stop SARS-CoV-2 spreading, and also to preserve RAS system and inhibit ROS generation by NOX [28]. Interestingly, in an in vitro and in vivo study, NOX4-derived ROS production was demonstrated to be modulated by ACE2 [193].
A new in vitro study demonstrated that the fusion protein of recombinant human [rh] ACE2 with a Fc fragment showed high affinity binding to the RBD of SARS-CoV-2 and potently neutralized SARS-CoV-2 entry [194]. In addition, a recent paper strongly supported the efficacy of rhACE2 against SARS-CoV-2 infection [195]. In particular, the authors reported that clinical-grade rh soluble ACE2 exhibited strong inhibitory activity against SARS-CoV-2 in cell cultures and in human blood vessels and kidney engineered copies [184]. Very interestingly, in a recent case report, Zoufaly et al. [196] found that the delivery of rhACE2 in a SARS-CoV-2 patient caused a marked clinical improvement associated with reduction of inflammatory markers and of Ang II with a striking rise of Ang 1–7 and Ang 1–9. Intriguingly, SARS-CoV-2 viremia was significantly reduced after the first day of administration and thereafter it remained undetectable [196].

5.4. Inhibitors of NLRP3 Inflammasome

Given the strong inflammatory potential of NLRP3 inflammasome in the pathogenesis of different inflammatory diseases, many efforts have been made in the last few years in the search of NLRP3 inhibitors. As recently reviewed [197], many natural products and pharmaceutical drugs have been identified as NLRP3 inhibitors. Among natural and pharmaceutical products, oridonin (derived from Rabdosia rubescens plant) and parthenolide (derived from feverfew plant) as well as Bay 11-7082 have been reported to strongly suppress NLRP3 inflammasome in experimental models [198,199]. Besides inhibiting NLRP3, parthenolide and Bay 11-7082 have also been shown to lower NF-kB activation and to prevent lung inflammation in animals affected by SARS-CoV [199]. Another drug reducing NLRP3 inflammasome activity and IL-1beta secretion in cells infected with RNA viruses is glyburide, a sulfonylurea extensively used in the treatment of type 2 diabetes [200,201]. Likewise, tranilast, a drug used for allergic conditions, was shown to reduce NF-kB activation and NLRP3 assembly in animal models of inflammatory diseases [202]. Similarly, colchicine, a drug used in autoinflammatory diseases for its effect of preventing adhesion and recruitment of neutrophils at endothelial surface [203], can also suppress NLRP3 inflammasome and production of IL-1beta and IL-18 [204]. Finally, mefenamic acid and flufenamic acid, belonging to the group of non-steroidal anti-inflammatory drugs, by inhibiting NLRP3 inflammasome and IL-1beta secretion, have been reported to strongly suppress viral replication independent of their cyclooxygenase-1 mediated anti-inflammatory activity [205,206]. Because of the key role of NLRP3 inflammasome activation in the pathogenesis of SARS-CoVs diseases and the promising results obtained by inhibitors of the NLRP3 inflammasome in pre-clinical and/or clinical studies [197], it can be hypothesized that its inhibition may potentially decrease tissue inflammation also in COVID-19.

5.5. Glucocorticoids (GCs) and Non-Steroidal Anti-Inflammatory Drugs (NSAIDs)

It is well established that GCs control inflammation through pleiotropic mechanisms [207,208]. In particular, GCs block the activation of transcription factors that mediate inflammatory responses, such as NF-kB and activator protein 1 [207], thus suppressing the synthesis of many pro-inflammatory cytokines and of inducible nitric oxide synthase. [207,209]. Furthermore, GCs reduce cyclooxygenase 2 activity by inducing the NF-kB inhibitor glucocorticoid-induced leucine zipper protein, thus weakening prostaglandin release [210]. GCs also inhibit adhesion molecule expression in endothelia cells and integrins in immune cells so diminishing leukocyte recruitment [207,211,212]. In addition, GCs reverse macrophages to an anti-inflammatory state, promote resolution of inflammation, and weaken antigen presentation in dendritic cells [207,213,214,215]. In view of this formidable strength, GCs are considered the cornerstone of the anti-inflammatory and immunosuppressive therapy. At the beginning of COVID-19 pandemic there were many perplexities for handling an infectious disease with potent immunosuppressive agents like GCs. Then, on the basis of the promising results derived from case reports and small observational studies, a series of large-scale randomized clinical trials were started. In the Randomised Evaluation of COVID-19 Therapy (RECOVERY, NCT04381936) trial [216], patients (n. 2104) were randomly assigned to receive oral or intravenous dexamethasone (6 mg once daily) for up to 10 days or to receive usual care (n. 4321). The preliminary results showed that in COVID-19 hospitalized patients, the use of dexamethasone significantly reduced 28-day mortality among those who were receiving either invasive mechanical ventilation or oxygen alone. On the contrary, dexamethasone had no effect among patients not requiring respiratory support. In the prospective meta-analysis of the Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group of the World Health Organization (WHO) [217], the authors analyzed pooled data from seven randomized clinical trials evaluating the efficacy of GCs in 1703 critically ill patients with COVID-19. Similarly to the RECOVERY study [216], the REACT study demonstrated that low-dose dexamethasone, compared with usual care or placebo, reduced all-cause mortality in hospitalized patients with COVID-19 who required respiratory support. Following these studies, the WHO released two recommendations establishing that GCs (dexamethasone per os or intravenously and hydrocortisone intravenously) should be given for 7 to 10 days only in critical and severe COVID-19 patients.
As for NSAIDs, it has been established that they operate by suppressing cyclooxygenase (COX) 1 and 2, thus limiting the synthesis of prostaglandins, which play a crucial role in the pathogenesis of fever and inflammation [218]. NSAIDs are habitually employed in SARS-CoV-2 infection to lower fever and alleviate muscle pain, but whether NSAIDs are helpful or damaging to COVID-19 patients is currently uncertain, and a cautious strategy is suggested [219,220,221]. Available data so far on the effects of chronic treatment with NSAIDs on SARS-CoV-2 infection are few, and have not been derived from randomized clinical trials. In particular, a large case control study showed that chronic treatment with NSAIDs was not associated with risk of COVID-19 infection or COVID-19 severity [222]. Similar results stem from a recent multicenter retrospective cohort study of hospitalized patients with COVID-19 demonstrating a lack of association between the pre-hospital use of NSAIDs and mortality [223]. Additionally, in a particular setting of COVID-19 patients with chronic inflammatory rheumatic disease, the prior treatment with NSAIDs did not influence the risk of hospitalization [224]. Concerning the potential role of NSAIDs as adjuvant therapy in COVID-19 patients, a recent pilot study showed that adjuvant treatment with celecoxib, a selective inhibitor of COX2, improved the recovery in non-severe and severe cases of SARS-CoV-2 patients and impeded the evolution to a critical step [225].
Although the WHO, the European Medicines Agency (EMA) and the United Kingdom National Health Service have stated that there is currently no scientific evidence that NSAIDs augment the risk or worsen SARS-CoV-2 infection, and that there is no reason for patients who are taking NSAIDs for chronic diseases to stop taking them, from a clinical point of view, it is now recommended that patients with COVID-19 should use paracetamol rather than NSAIDs [226]. This advice is further strengthened by previous clinical trials in non-SARS-CoV-2 pulmonary infectious diseases that have suggested avoiding these drugs (reviewed in [227]).

6. Conclusions

The redox-regulated intracellular pathways triggered and used by viruses may constitute a new and encouraging target for novel approaches in the control and therapy of viral infections. In this context, it has been demonstrated that respiratory viral infections and in particular SARS-CoV-2, despite a dysregulation of ROS production, inhibit NRF2 and activate NF-kB pathways, resulting in inflammation and oxidative injury. The outstanding results from experimental studies available so far clearly indicate the need to also test NRF2 activators in randomized clinical trials in patients with SARS-CoV-2 infection.

Author Contributions

Conceptualization, A.M.F.P. and C.M.; writing—original draft preparation, L.C., C.S., A.M.F.P.; writing—review and editing, C.M. and A.M.F.P. All The authors have approved the submitted version and agree to be personally accountable for the author’s own contributions and for ensuring that questions related to the accuracy or integrity of any part of the work, even ones in which the author was not personally involved, are appropriately investigated, resolved, and documented in the literature. All authors have read and agreed to the published version of the manuscript.

Funding

This manuscript received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACEAngiotensin Converting Enzyme
AngAngiotensin
AT1RAngiotensin II Type-1 Receptor
ADAM17A Disintegrin And Metalloproteinase17
COXcyclooxygenase
COVID-19Coronavirus disease 2019
DAMPsdamage-associated molecular patterns
DMFDimethyl Fumarate
EMAEuropean Medicines Agency
FiO2Fraction Of Inspired Oxygen
FPFusion protein
GCsGlucocorticoids
GSHGlutathione
HOHeme-Oxygenase
HbHemoglobin
Keap-1Kelch-like enoyl-CoA hydratase-associated protein 1
IFNInterferon
ILInterleukin
MAVSMitochondrial antiviral signaling system;
MCP1monocyte chemoattractant protein-1
MEAs-acetyl-mercaptoethylamine
MERSMiddle East respiratory syndrome
NACN-acetylcysteine
NETsNeutrophil extracellular traps
NLRP3NOD-like receptors protein 3
NRP1Neuropilin-1
NOXNicotinamide adenine dinucleotide phosphate (NADPH) oxidases
NF-kBNuclear Factor-κB
NRF2Nuclear factor erythroid 2 p45-related factor2
NSAIDsnon-steroidal anti-inflammatory drugs
4-OI4-Octyl-Itaconate
Orf9bOpen reading frame-9b
OSOxidative stress
OxPLsOxidized phospholipids
PAI-1Plasminogen Activator Inhibitor-1
PAMPsPathogen-associated molecular patterns
RASRenin-angiotensin system
RBDReceptor-binding domain
rhRecombinant Human
ROSReactive oxygen species
SARS-CoV-2Severe Acute Respiratory Syndrome (SARS)-like Coronavirus
S proteinViral Membrane Spike protein
S1Receptor-binding subunit
S2Membrane fusion subunit
scRNA-seqsingle cell RNA-sequence
TRXIPThioredoxin Interacting/Inhibiting Protein;
TOMTranslocase of Outer Membrane
TMPRSS2Transmembrane protease serine 2
WHOWorld Health Organization

References

  1. Li, Q.; Guan, X.; Wu, P.; Wang, X.; Zhou, L.; Tong, Y.; Ren, R.; Leung, K.S.M.; Lau, E.H.Y.; Wong, J.Y.; et al. Early transmission dynamics in Wuhan, China, of novel coronavirus infected pneumonia. N. Engl. J. Med. 2020, 382, 1199–1207. [Google Scholar] [CrossRef]
  2. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  3. Qamar, M.T.; Alqahtani, S.M.; Alamri, M.A.; Chen, L.L. Structural basis of SARSCoV-2 3CLpro and anti-COVID-19 drug discovery from medicinal plants. J. Pharm. Anal. 2020, 4, 313–319. [Google Scholar] [CrossRef]
  4. Chan, J.W.; Ng, C.K.; Chan, Y.H.; Mok, T.Y.; Lee, S.; Chu, S.Y.; Law, W.L.; Lee, M.P.; Li, P.C. Short term outcome and risk factors for adverse clinical outcomes in adults with severe acute respiratory syndrome [SARS]. Thorax 2003, 58, 686–689. [Google Scholar] [CrossRef] [Green Version]
  5. Badawi, A.; Ryoo, S.G. Prevalence of comorbidities in the Middle East respiratory syndrome coronavirus [MERS-CoV]: A systematic review and meta-analysis. Int. J. Infect. Dis. 2016, 49, 129–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
  7. Liu, K.; Fang, Y.Y.; Deng, Y.; Liu, W.; Wang, M.F.; Ma, J.P.; Xiao, W.; Wang, Y.N.; Zhong, M.H.; Li, C.H.; et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin. Med. J. 2020, 133, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
  9. Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
  10. Han, H.; Xie, L.; Liu, R.; Yang, J.; Liu, F.; Wu, K.; Chen, L.; Hou, W.; Feng, Y.; Zhu, C. Analysis of heart injury laboratory parameters in 273 COVID-19 patients in one hospital in Wuhan, China. J. Med. Virol. 2020, 92, 819–823. [Google Scholar] [CrossRef] [PubMed]
  11. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult in patients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
  12. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.; et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020, 82, 1708–1720. [Google Scholar] [CrossRef]
  13. Li, Z.; Wu, M.; Yao, J.; Guo, J.; Liao, X.; Song, S.; Li, J.; Duan, G.; Zhou, Y.; Wu, X.; et al. Caution on kidney dysfunctions of COVID-19 patients. medRxiv 2020. [Google Scholar] [CrossRef]
  14. Mao, L.; Wang, M.; Chen, S.; He, Q.; Chang, J.; Hong, C.; Zhou, Y.; Wang, D.; Li, Y.; Jin, H.; et al. Neurologic manifestations of hospitalized patients with COVID-19 in Wuhan, China: A retrospective case series study. JAMA Neurol. 2020, 77, 683–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wu, Y.; Xu, X.; Chen, Z.; Duan, J.; Hashimoto, K.; Yang, L.; Liu, C.; Yang, C. Nervous system involvement after infection with COVID-19 and other coronaviruses. Brain Behav. Immun. 2020, 87, 18–22. [Google Scholar] [CrossRef]
  16. Yao, X.H.; Li, T.Y.; He, Z.C.; Ping, Y.F.; Liu, H.W.; Yu, S.C.; Mou, H.M.; Wang, L.H.; Zhang, H.R.; Fu, W.J. A pathological report of three COVID-19 cases by minimally invasive autopsies. Zhonghua Bing Li Xue Za Zhi 2020, 8, 411–417. [Google Scholar]
  17. Xie, H.; Zhao, J.; Lian, N.; Lin, S.; Xie, Q.; Zhuo, H. Clinical characteristics of non-ICU hospitalized patients with coronavirus disease 2019 and liver injury: A Retrospective study. Liver Int. 2020, 40, 1321–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Zhang, Y.; Zheng, L.; Liu, L.; Zhao, M.; Xiao, J.; Zhao, Q. Liver impairment in COVID-19 patients: A retrospective analysis of 115 cases from a single center in Wuhan city, China. Liver Int. 2020, 40, 2095–2103. [Google Scholar] [CrossRef] [Green Version]
  19. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H. Clinical and immunologic features in severe and moderate Coronavirus Disease 2019. J. Clin. Investig. 2020, 30, 2620–2629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef]
  21. Han, H.; Yang, L.; Liu, R.; Liu, F.; Wu, K.L.; Li, J.; Liu, X.H.; Zhu, C.L. Prominent changes in blood coagulation of patients with SARS-CoV-2 infection. Clin. Chem. Lab. Med. 2020, 58, 1116–1120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef] [Green Version]
  23. Coronavirus WHO. COVID-19. WHO. 2020. Available online: https://who.sprinklr.com/ (accessed on 31 December 2020).
  24. Li, Y.; Zhou, W.; Yang, L.; You, R. Physiological and pathological regulation of ACE2, the SARS-CoV-2 receptor. Pharmacol. Res. 2020, 157, 104833. [Google Scholar] [CrossRef] [PubMed]
  25. Wan, Y.; Shang, J.; Graham, R.; Baric, R.S.; Li, F. Receptor recognition by novel coronavirus from Wuhan: An analysis based on decade-long structural studies of SARS. J. Virol. 2020, 94, e00127-20. [Google Scholar] [CrossRef] [Green Version]
  26. Kuhn, J.H.; Li, W.; Choe, H.; Farzan, M. Angiotensin-converting enzyme 2: A functional receptor for SARS coronavirus. Cell. Mol. Life Sci. 2004, 61, 2738–2743. [Google Scholar] [CrossRef] [PubMed]
  27. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef] [Green Version]
  28. Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W.; et al. A crucial role of angiotensin converting enzyme 2 ACE2 in SARS coronavirus-induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef] [PubMed]
  29. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.M.; Wang, W.; Song, Z.G.; Hu, Y.; Tao, Z.W.; Tian, J.H.; Pei, Y.Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269. [Google Scholar] [CrossRef] [Green Version]
  30. Jiang, S.; Hillyer, C.; Du, L. Neutralizing antibodies against SARS-CoV-2 and other human Coronaviruses. Trends Immunol. 2020, 41, 355–359. [Google Scholar] [CrossRef]
  31. Shang, J.; Wana, Y.; Luoa, C.; Yea, G.; Genga, Q.; Auerbacha, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  32. Walls, A.C.; Park, Y.J.; Tortorici, M.A.; Wall, A.; McGuire, A.T.; Veesler, D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 2020, 181, 281–292. [Google Scholar] [CrossRef]
  33. Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; et al. Structure of the SARS-CoV-2 spike receptor binding domain bound to the ACE2 receptor. Nature 2020, 581, 215–220. [Google Scholar] [CrossRef] [Green Version]
  34. Shang, J.; Ye, G.; Shi, K.; Wan, Y.; Luo, C.; Aihara, H.; Geng, Q.; Auerbach, A.; Li, F. Structural basis of receptor recognition by SARS-CoV-2. Nature 2020, 581, 221–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yuan, Y.; Cao, D.; Zhang, Y.; Ma, J.; Qi, J.; Wang, Q.; Lu, G.; Wu, Y.; Yan, J.; Shi, Y.; et al. Cryo-EM structures of MERS-CoV and SARS-CoV spike glycoproteins reveal the dynamic receptor binding domains. Nat. Commun. 2017, 8, 15092. [Google Scholar] [CrossRef] [PubMed]
  36. Cantuti-Castelvetri, L.; Ojha, R.; Pedro, L.D.; Djannatian, M.; Franz, J.; Kuivanen, S.; van der Meer, F.; Kallio, K.; Kaya, T.; Anastasina, M.; et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 2020, 370, 856–860. [Google Scholar] [CrossRef]
  37. Daly, J.L.; Simonetti, B.; Klein, K.; Chen, K.E.; Williamson, M.K.; Antón-Plágaro, C.; Deborah, K.; Shoemark, D.K.; Simón-Gracia, L.; Bauer, M.; et al. Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science 2020, 370, 861–865. [Google Scholar] [CrossRef]
  38. Chen, Y.; Liu, Q.; Guo, D.J. Emerging coronaviruses: Genome structure, replication, and pathogenesis. Med. Virol. 2020, 92, 418–423. [Google Scholar] [CrossRef]
  39. Chappell, M.C. Biochemical evaluation of the renin-angiotensin system: Then good, bad, and absolute? Am. J. Physiol. 2016, 310, 137–152. [Google Scholar] [CrossRef] [Green Version]
  40. Erdos, E.G. Conversion of angiotensin I to angiotensin II. Amer. J. Med. 1976, 60, 749–759. [Google Scholar] [CrossRef]
  41. Tikellis, C.; Thomas, M.C. Angiotensin-Converting Enzyme 2 [ACE2] is a key modulator of the Renin Angiotensin System in health and disease. Int. J. Pept. 2012, 25, 62–94. [Google Scholar] [CrossRef] [PubMed]
  42. Towler, P.; Staker, B.; Prasad, S.G.; Menon, S.; Tang, J.; Parsons, T.; Ryan, D.; Fisher, M.; Williams, D.; Natalie, A. ACE2 X-ray structures reveal a large hinge-bending motion important for inhibitor binding and catalysis. J. Biol. Chem. 2004, 279, 17996–18007. [Google Scholar] [CrossRef] [Green Version]
  43. Wang, K.; Gheblawi, M.; Oudit, G.Y. Angiotensin converting enzyme 2: A double-edged sword. Circulation 2020, 142, 426–428. [Google Scholar] [CrossRef]
  44. Gheblawi, M.; Wang, K.; Viveiros, A.; Nguyen, Q.; Zhong, J.C.; Turner, A.J.; Raizada, M.K.; Grant, M.B.; Oudit, G.Y. Angiotensin-Converting Enzyme 2: SARS-CoV-2 Receptor and Regulator of the Renin-Angiotensin System: Celebrating the 20th Anniversary of the Discovery of ACE2. Circ. Res. 2020, 126, 1456–1474. [Google Scholar] [CrossRef]
  45. Burrell, L.M.; Johnston, C.I.; Tikellis, C.; Cooper, M.E. ACE2, a new regulator of the renin-angiotensin system. Trends Endocrinol. Metab. 2004, 15, 166–169. [Google Scholar] [CrossRef]
  46. Hikmet, F.; Méar, L.; Edvinsson, A.; Micke, P.; Uhlén, M.; Lindskog, C. The protein expression profile of ACE2 in human tissues. Mol. Syst. Biol. 2020, 16, e9610. [Google Scholar] [CrossRef]
  47. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Datta, P.K.; Liu, F.; Fischer, T.; Rappaport, J.; Qin, X. SARS-CoV-2 pandemic and research gaps: Understanding SARS-CoV-2 interaction with the ACE2 receptor and implications for therapy. Theranostics 2020, 10, 7448–7464. [Google Scholar] [CrossRef] [PubMed]
  49. Liu, Y.; Yang, Y.; Zhang, C.; Huang, F.; Wang, F.; Yuan, J.; Wang, Z.; Li, J.; Li, J.; Feng, C.; et al. Clinical and biochemical indexes from 2019-nCoV infected patients linked to viral loads and lung injury. Sci. China Life Sci. 2020, 63, 364–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Patel, V.B.; Clarke, N.; Wang, Z.; Fan, D.; Parajuli, N.; Basu, R.; Putko, B.; Kassiri, Z.; Turner, A.J.; Oudit, G.Y. Angiotensin II induced proteolytic cleavage of myocardial ACE2 is mediated by TACE/ADAM-17: A positive feedback mechanism in the RAS. J. Mol. Cell. Cardiol. 2014, 66, 167–176. [Google Scholar] [CrossRef] [PubMed]
  51. Xu, P.; Derynck, R. Direct activation of TACE-mediated ectodomain shedding by p38 MAP kinase regulates EGF receptor-dependent cell proliferation. Mol. Cell. 2010, 37, 551–566. [Google Scholar] [CrossRef] [Green Version]
  52. Black, R.A.; Rauch, C.T.; Kozlosky, C.J.; Peschon, J.J.; Slack, J.L.; Wolfson, M.F.; Castner, B.J.; Stocking, K.L.; Reddy, P.; Srinivasan, S.; et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature 1997, 385, 729–733. [Google Scholar] [CrossRef]
  53. Bzowska, M.; Jura, N.; Lassak, A.; Black, R.A.; Bereta, J. Tumour necrosis factor-alpha stimulates expression of TNF-alpha converting enzyme in endothelial cells. Eur. J. Biochem. 2004, 271, 2808–2820. [Google Scholar] [CrossRef]
  54. Zablocki, D.; Sadoshima, J. Angiotensin II and Oxidative Stress in the Failing Heart. Antioxid. Redox Signal. 2013, 19, 1095–1109. [Google Scholar] [CrossRef]
  55. Dikalov, S.I.; Nazarewicz, R.R. Angiotensin II-Induced Production of Mitochondrial Reactive Oxygen Species: Potential Mechanisms and Relevance for Cardiovascular Disease. Antioxid. Redox Signal. 2013, 19, 1085–1094. [Google Scholar] [CrossRef]
  56. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Kruger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280. [Google Scholar] [CrossRef] [PubMed]
  57. Vaarala, M.H.; Porvari, K.S.; Kellokumpu, S.; Kyllönen, A.P.; Vihko, P.T. Expression of transmembrane serine protease TMPRSS2 in mouse and human tissues. J. Pathol. 2001, 193, 134–140. [Google Scholar] [CrossRef]
  58. Metzger, E.; Willmann, D.; McMillan, J.; Forne, I.; Metzger, P.; Gerhardt, S.; Petroll, K.; von Maessenhausen, A.; Urban, S.; Schott, A.K.; et al. Assembly of methylated KDM1A and CHD1 drives androgen receptor-dependent transcription and translocation. Nat. Struct. Mol. Biol. 2016, 23, 132–139. [Google Scholar] [CrossRef] [PubMed]
  59. Lukassen, S.; Chua, R.L.; Trefzer, T.; Kahn, N.C.; Schneider, M.; Muley, T.; Winter, H.; Meister, M.; Veith, C.; Boots, A.W.; et al. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J. 2020, 39, e105114. [Google Scholar] [CrossRef] [PubMed]
  60. Wrapp, N.D.; Wang, K.S.; Corbett, J.A.; Goldsmith, C.-L.; Hsieh, O.; Abiona, B.S.; Graham, J.S. McLellan, Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260–1263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Hoffmann, M.; Kleine-Weber, H.; Pöhlmann, S. A multibasic cleavage site in the spike protein of SARS-CoV-2 is essential for infection of human lung cells. Mol. Cell 2020, 78, 779–784. [Google Scholar] [CrossRef]
  62. Lin, B.; Ferguson, C.; White, J.T.; Wang, S.; Vessella, R.; True, L.D.; Hood, L.; Nelson, P.S. Prostate-localized and androgen-regulated expression of the membrane bound serine protease TMPRSS2. Cancer Res. 1999, 59, 4180–4184. [Google Scholar]
  63. Yoo, S.; Pettersson, A.; Jordahl, K.M.; Lis, R.T.; Lindstrom, S.; Meisner, A.; Nuttall, E.J.; Stack, E.C.; Stampfer, M.J.; Kraft, K.; et al. Androgen receptor CAG repeat polymorphism and risk of TMPRSS2:ERG-positive prostate cancer. Cancer Epidemiol. Biomarkers Prev. 2014, 23, 2027–2031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Mikkonen, L.; Pihlajamaa, P.; Sahu, B.; Zhang, F.P.; Janne, O.A. Androgen receptor and androgen-dependent gene expression in lung. Mol. Cell Endocrinol. 2010, 317, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Richardson, S.; Hirsch, J.S.; Narasimhan, M.; Crawford, J.M.; McGinn, T.; Davidson, K.W.; Barnaby, D.P.; Becker, L.B.; Chelico, J.D.; Cohen, S.L.; et al. Presenting Characteristics, Comorbidities, and Outcomes among 5700 Patients Hospitalized with COVID-19 in the New York City Area. JAMA 2020, 323, 2052–2059. [Google Scholar] [CrossRef]
  66. Onder, G.; Rezza, G.; Brusaferro, S. Case-Fatality Rate and Characteristics of Patients Dying in Relation to COVID-19 in Italy. JAMA 2020, 323, 1775–1776. [Google Scholar] [CrossRef] [PubMed]
  67. Cattrini, C.; Bersanelli, M.; Latocca, M.M.; Conte, B.; Vallome, G.; Boccardo, F. Sex Hormones and Hormone Therapy during COVID-19 Pandemic: Implications for Patients with Cancer. Cancers 2020, 12, 2325. [Google Scholar] [CrossRef] [PubMed]
  68. Delgado-Roche, L.; Mesta, F. Oxidative stress as key player in severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) infection. Arch. Med. Res. 2020, 51, 384–387. [Google Scholar] [CrossRef]
  69. Khomich, O.A.; Kochetkov, S.N.; Bartosch, B.; Ivanov, A. Redox Biology of Respiratory viral infections. Viruses 2018, 10, 392. [Google Scholar] [CrossRef] [Green Version]
  70. Checconi, P.; De Angelis, M.; Marcocci, M.E.; Fraternale, A.; Magnani, M.; Palamara, A.T.; Nencioni, L. Redox-modulating agents in the treatment of viral Infections. Int. J. Mol. Sci. 2020, 21, 4084. [Google Scholar] [CrossRef]
  71. Cecchini, R.; Cecchini, A.L. SARS-CoV-2 infection pathogenesis is related to oxidative stress as a response to aggression. Med. Hypotheses 2020, 143, 101–102. [Google Scholar] [CrossRef] [PubMed]
  72. Merad, M.; Martin, J.C. Pathological inflammation in patients with COVID-19: A key role for monocytes and macrophages. Nat. Rev. Immunol. 2020, 20, 355–362. [Google Scholar] [CrossRef] [PubMed]
  73. Chernyak, B.V.; Popova, E.N.; Prikhodko, A.S.; Grebenchikov, O.A.; Zinovkina, L.A.; Zinovkin, R.A. COVID-19 and oxidative stress. Biochemistry 2020, 85, 1543–1553. [Google Scholar]
  74. Fernandes, I.G.; de Brito, C.A.; Dos Reis, V.M.S.; Sato, M.N.; Pereira, N.Z. SARS-CoV-2 and other respiratory viruses: What does oxidative Stress have to do with it? Oxid. Med. Cell. Longev. 2020, 2020, 8844280. [Google Scholar] [CrossRef] [PubMed]
  75. Suhail, S.; Zajac, J.; Fossum, C.; Lowater, H.; McCracken, C.; Severson, N.; Laatsch, B.; Narkiewicz-Jodko, A.; Johnson, B.; Liebau, J.; et al. Role of oxidative Stress on SARS-CoV (SARS) and SARS-CoV-2 (COVID-19) Infection: A review. Protein J. 2020, 39, 644–656. [Google Scholar] [CrossRef]
  76. Violi, F.; Oliva, A.; Cangemi, R.; Ceccarelli, G.; Pignatelli, P.; Carnevale, R.; Cammisotto, V.; Lichtner, M.; Alessandri, F.; De Angelis, M.; et al. Nox2 activation in Covid-19. Redox Biol. 2020, 36, 101655. [Google Scholar] [CrossRef]
  77. Libby, P.; Luscher, T. COVID-19 is, in the end, an endothelial disease. Eur. Heart J. 2020, 41, 3038–3044. [Google Scholar] [CrossRef]
  78. Imai, Y.; Kuba, K.; Neely, G.G.; Yaghubian-Malhami, R.; Perkmann, T.; van Loo, G.; Ermolaeva, M.; Veldhuizen, R.; Leung, Y.H.; Wang, H.; et al. Identification of oxidative stress and Toll- like receptor 4 signaling as a key pathway of acute lung injury. Cell 2008, 133, 235–249. [Google Scholar] [CrossRef]
  79. Berliner, J.A.; Watson, A.D. A role for oxidized phospholipids in atherosclerosis. N. Engl. J. Med. 2005, 353, 9–11. [Google Scholar] [CrossRef]
  80. Owens, A.P.; Passam, F.H.; Antoniak, S.; Marshall, S.M.; McDaniel, A.L.; Rudel, L.; Williams, J.C.; Hubbard, B.K.; Dutton, J.A.; Wang, J.; et al. Monocyte tissue factor-dependent activation of coagulation in hypercholesterolemic mice and monkeys is inhibited by simvastatin. J. Clin. Investig. 2012, 122, 558–568. [Google Scholar]
  81. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tan, D. Ferroptosis: Process and function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [Green Version]
  82. Yang, M.; Lai, C.L. SARS-CoV-2 infection: Can ferroptosis be a potential treatment target for multiple organ involvement. Cell Death Discov. 2020, 6, 130. [Google Scholar] [CrossRef] [PubMed]
  83. Edeas, M.; Saleh, J.; Peyssonnaux, C. Iron: Innocent bystander or vicious culprit in COVID-19 pathogenesis? Int. J. Infect. Dis. 2020, 97, 303–305. [Google Scholar] [CrossRef]
  84. Lin, C.W.; Lin, K.H.; Hsieh, T.H.; Shiu, S.Y.; Li, J.Y. Severe acute respiratory syndrome coronavirus 3C-like protease-induced apoptosis. FEMS Immunol. Med. Microbiol. 2006, 46, 375–380. [Google Scholar] [CrossRef] [Green Version]
  85. Collins, Y.; Chouchani, E.T.; James, A.M.; Menger, K.E.; Cocheme, H.M.; Murphy, M.P. Mitochondrial redox signalling at a glance. J. Cell Sci. 2012, 125, 801–806. [Google Scholar] [CrossRef] [Green Version]
  86. Dunn, J.D.; Alvarez, L.A.; Zhang, X.; Soldati, T. Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol. 2015, 6, 472–485. [Google Scholar] [CrossRef]
  87. Anand, S.K.; Tikoo, S.K. Viruses as modulators of mitochondrial functions. Adv. Virol. 2013, 20, 738794. [Google Scholar] [CrossRef]
  88. de las Heras, N.; Giménez, V.M.M.; Ferder, L.; Manucha, W.; Lahera, V. Implications of Oxidative Stress and Potential Role of Mitochondrial Dysfunction in COVID-19: Therapeutic Efects of Vitamin D. Antioxidants 2020, 9, 897. [Google Scholar] [CrossRef]
  89. Burtscher, J.; Cappellano, G.; Omori, A.; Koshiba, T.; Millet, G.P. Mitochondria: In the Cross fire of SARS-CoV-2 and immunity. Science 2020, 23, 101631. [Google Scholar]
  90. Shi, C.S.; Qi, H.Y.; Boularan, C.; Huang, N.N.; Abu-Asab, M.; Shelhamer, J.H.; Kehrl, J.H. SARS-CoV ORF-9b suppresses innate immunity by targeting mitochondria and the MAVS/TRAF3/TRAF6 signalosome. J. Immunol. 2014, 193, 3080–3089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. McBride, R.; Fielding, B.C. The role of severe acute respiratory syndrome (SARS)-coronavirus accessory proteins in virus pathogenesis. Viruses 2013, 4, 2902–2923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Scherz-Shouval, R.; Elazar, Z. Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem. Sci. 2011, 36, 30–38. [Google Scholar] [CrossRef] [PubMed]
  93. Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; White, K.M.; O’Meara, M.J.; Rezelj, V.V.; Guo, J.Z.; Danielle, L. A SARS-CoV-2 protein interaction map reveals targets for drug-repurposing. Nature 2020, 583, 459–468. [Google Scholar] [CrossRef] [PubMed]
  94. Jiang, H.; Zhang, H.; Meng, Q.; Xie, J.; Li, Y.; Chen, H.; Zheng, Y.; Wang, X.; Qi, H.; Zhang, J.; et al. SARS-CoV-2 Orf9b suppresses type I interferon responses by targeting TOM70. Cell. Mol. Immunol. 2020, 17, 998–1000. [Google Scholar] [CrossRef]
  95. Filadi, R.; Leal, N.S.; Schreiner, B.; Rossi, A.; Dentoni, G.; Pinho, C.M.; Wiehager, B.; Cieri, D.; Calì, T.; Pizzo, P. TOM70 sustains cell bioenergetics by promoting IP3R3-Mediated ER to mitochondria Ca(2+) transfer. Curr. Biol. 2018, 28, 369–382. [Google Scholar] [CrossRef] [Green Version]
  96. Sgarbanti, R.; Nencioni, L.; Amatore, D.; Coluccio, P.; Fraternale, A.; Sale, P.; Mammola, C.L.; Carpino, G.; Gaudio, E.; Magnani, M.; et al. Redox regulation of the influenza hemagglutinin maturation process: A new cell-mediated strategy for anti-influenza therapy. Antioxid. Redox Signal. 2011, 15, 593–606. [Google Scholar] [CrossRef]
  97. Cao, M.; Zhang, D.; Wang, Y.; Lu, Y.; Zhu, X.; Li, Y.; Xue, H.; Lin, Y.; Zhang, M.; Sun, Y.; et al. Clinical features of patients infected with the 2019 novel coronavirus [COVID-19] in Shanghai, China. medRxiv 2020. preprint. [Google Scholar] [CrossRef] [Green Version]
  98. Silvagno, F.; Vernone, A.; Pescarmona, G.P. The Role of Glutathione in Protecting against the Severe Inflammatory Response Triggered by COVID-19. Antioxidants 2020, 9, 624. [Google Scholar] [CrossRef] [PubMed]
  99. Wu, C.; Chen, X.; Cai, Y.; Xia, J.; Zhou, X.; Xu, S.; Huang, H.; Zhang, L.; Zhou, X.; Du, C.; et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern. Med. 2020, 180, 934–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Ruan, Q.; Yang, K.; Wang, W.; Jiang, L.; Song, J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020, 46, 846–848. [Google Scholar] [CrossRef] [Green Version]
  101. Gong, J.; Dong, H.; Xia, Q.; Huang, Z.; Wang, D.; Zhao, Y.; Liu, W.; Tu, S.; Zhang, M.; Wang, Q.; et al. Correlation analysis between disease severity and inflammation-related parameters in patients with COVID-19 pneumonia. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  102. Martinon, F. Detection of immune danger signals by NALP3. J. Leukoc. Biol. 2008, 83, 507–511. [Google Scholar] [CrossRef] [PubMed]
  103. Siu, K.; Yuen, K.; Castano-Rodriguez, C.; Ye, Z.; Yeung, M.; Fung, S.; Yuan, S.; Chan, C.; Yuen, K.; Enjuanes, L.; et al. Severe acute respiratory syndrome Coronavirus ORF3a protein activates the NLRP3 inflammasome by promoting TRAF3-dependent ubiquitination of ASC. FASEB J. 2019, 33, 8865–8877. [Google Scholar] [CrossRef] [PubMed]
  104. Broz, P.; Dixit, V.M. Inflammasomes: Mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 2016, 16, 407–420. [Google Scholar] [CrossRef] [PubMed]
  105. Fox, S.E.; Akmatbekov, A.; Harbert, J.L.; Li, G.; Brown, J.Q.; Vander Heide, R.S. Pulmonary and cardiac pathology in Covid-19: The first autopsy series from New Orleans. medRxiv 2020. [Google Scholar] [CrossRef]
  106. Tian, S.; Hu, W.; Niu, L.; Liu, H.; Xu, H.; Xiao, S.Y. Pulmonary pathology of early phase 2019 novel coronavirus [COVID-19] pneumonia in two patients with lung cancer. J. Thorac. Oncol. 2020, 15, 700–704. [Google Scholar] [CrossRef]
  107. Yang, M. Cell pyroptosis, a potential pathogenic mechanism of 2019-nCoV Infection, SSRN. Electron. J. 2020, 3527420. [Google Scholar]
  108. Cookson, B.T.; Brennan, M.A. Pro-inflammatory programmed cell death. Trends Microbiol. 2001, 9, 113–114. [Google Scholar] [CrossRef]
  109. Fink, S.L.; Cookson, B.T. Apoptosis, pyroptosis, and necrosis: Mechanistic description of dead and dying eukaryotic cells. Infect. Immun. 2005, 73, 1907–1916. [Google Scholar] [CrossRef] [Green Version]
  110. Faust, H.; Mangalmurti, N.S. Collateral damage: Necroptosis in the development of lung injury. Am. J. Phys. Lung Cell. Mol. Phys. 2020, 318, 215–225. [Google Scholar] [CrossRef]
  111. Sauler, M.; Bazan, I.S.; Lee, P.J. Cell death in the lung: The apoptosis-necroptosis axis. Annu. Rev. Physiol. 2019, 81, 375–402. [Google Scholar] [CrossRef]
  112. Ueno, H.; Matsuda, T.; Hashimoto, S.; Amaya, F.; Kitamura, Y.; Tanaka, M.; Kobayashi, A.; Maruyama, I.; Yamada, S.; Hasegawa, N.; et al. Contributions of high mobility group box protein in experimental and clinical acute lung injury. Am. J. Respir. Crit. Care Med. 2004, 170, 1310–1316. [Google Scholar]
  113. Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef] [PubMed]
  114. Mason, R.J. Pathogenesis of COVID-19 from a cell biologic perspective. Eur. Respir. J. 2020, 55, 2000607. [Google Scholar] [CrossRef] [Green Version]
  115. Aberdein, J.D.; Cole, J.; Bewley, M.A.; Marriott, H.M.; Dockrell, D.H. Alveolar macrophages in pulmonary host defence the unrecognized role of apoptosis as a mechanism of intracellular bacterial killing. Clin. Exp. Immunol. 2013, 174, 193–202. [Google Scholar]
  116. Losa García, J.E.; Rodríguez, F.M.; Martín de Cabo, M.R.; García Salgado, M.J.; Losada, J.P.; Villarón, L.G.; López, A.J.; Arellano, J.L. Evaluation of inflammatory cytokine secretion by human alveolar macrophages. Mediat. Inflamm. 1999, 8, 43–51. [Google Scholar] [CrossRef] [Green Version]
  117. Han, S.; Mallampalli, R.K. The acute respiratory distress syndrome: From mechanism to translation. J. Immunol. 2015, 194, 855–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Frantzeskaki, F.; Armaganidis, A.; Orfanos, S.E. Immunothrombosis in acute respiratory distress syndrome: Cross talks between inflammation and coagulation. Respiration 2017, 93, 212–225. [Google Scholar] [CrossRef] [PubMed]
  119. Haouari, M. Platelet oxidative stress and its relationship with cardiovascular diseases in type 2 diabetes mellitus patients. Curr. Med. Chem. 2019, 26, 4145–4165. [Google Scholar] [CrossRef]
  120. Freedman, J.E. Oxidative stress and platelets. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 11–16. [Google Scholar] [CrossRef] [Green Version]
  121. Øynebråten, I.; Barois, N.; Bergeland, T.; Küchler, A.M.; Bakke, O.; Haraldsen, G. Oligomerized, filamentous surface presentation of RANTES/CCL5 on vascular endothelial cells. Sci. Rep. 2015, 6, 9261. [Google Scholar] [CrossRef] [Green Version]
  122. Sonmez, O.; Sonmez, M. Role of platelets in immune system and inflammation. Porto Biomed. J. 2017, 2, 311–314. [Google Scholar] [CrossRef]
  123. Finsterbusch, M.; Schrottmaier, W.; Kral-Pointner, J.B.; Salzmann, M.; Asinger, A. Measuring and interpreting platelet-leukocyte aggregates. Platelets 2018, 29, 677–685. [Google Scholar] [CrossRef]
  124. Sreeramkumar, V.; Adrover, J.M.; Ballesteros, I.; Cuartero, M.I.; Rossaint, J.; Bilbao, I.; Nácher, M.; Pitaval, C.; Radovanovic, I.; Fukui, Y.; et al. Neutrophils scan for activated platelets to initiate inflammation. Science 2014, 346, 1234–1238. [Google Scholar] [CrossRef] [Green Version]
  125. Engelmann, B.; Massberg, S. Thrombosis as an intravascular effector of innate Immunity. Nat. Rev. Immunol. 2013, 13, 34–45. [Google Scholar] [CrossRef] [PubMed]
  126. Kimball, A.S.; Obi, A.T.; Diaz, J.A.; Henke, P.K. The emerging role of NETs in venous thrombosis and immunothrombosis. Front. Immunol. 2016, 7, 236. [Google Scholar] [CrossRef] [Green Version]
  127. Pfeiler, S.; Massberg, S.; Engelmann, B. Biological basis and pathological relevance of microvascular thrombosis. Thromb. Res. 2014, 133, 35–37. [Google Scholar] [CrossRef] [PubMed]
  128. Prabhakaran, P.; Ware, L.B.; White, K.E.; Cros, M.T.; Matthay, M.A.; Olman, M.A. Elevated levels of plasminogen activator inhibitor-1 in pulmonary edema fluid are associated with mortality in acute lung injury. Am. J. Phys. Lung Cell. Mol. Phys. 2003, 285, 20–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Sapru, A.; Curley, M.A.Q.; Brady, S.; Matthay, M.A.; Flori, H. Elevated PAI-1 is associated with poor clinical outcomes in pediatric patients with acute lung Injury. Intens. Care Med. 2010, 36, 157–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Xue, M.; Sun, Z.; Shao, M.; Yin, J.; Deng, Z.; Zhang, J.; Xing, L.; Yang, X.; Chen, B.L.; Dong, Z.; et al. Diagnostic and prognostic utility of tissue factor for severe sepsis and sepsis-induced acute lung injury. J. Transl. Med. 2015, 13, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Yadav, H.; Kor, D.J. Platelets in the pathogenesis of acute respiratory distress Syndrome. Amer. J. Physiol. Lung Cell. Mol. Physiol. 2015, 309, 915–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Gando, S.; Otomo, Y. Local hemostasis, immunothrombosis, and systemic disseminated intravascular coagulation in trauma and traumatic shock. Crit. Care 2015, 19, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Zuo, Y.; Yalavarthi, S.; Shi, H.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Weber, A.; Barnes, B.J.; Egeblad, M.; et al. Neutrophil extracellular traps in COVID-19. JCI Insight 2020, e138999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Gattinoni, L.; Coppola, S.; Cressoni, M.; Busana, M.; Rossi, S.; Chiumello, S.D. Covid-19 does not lead to a “typical” acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2020, 201, 1299–1300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Long, Y.; Liu, X.; Tan, X.Z.; Jiang, C.X.; Chen, S.W.; Liang, G.N.; He, X.M.; Wu, J.; Chen, T.; Xu, Y. ROS-induced NLRP3 inflammasome priming and activation mediate PCB 118-induced pyroptosis in endothelial cells. Ecotoxicol. Environ. Saf. 2020, 189, 109937. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, Y.; Shi, P.; Chen, Q.; Huang, Z.; Zou, D.; Zhang, J.; Gao, X.; Lin, Z. Mitochondrial ROS promote macrophage pyroptosis by inducing GSDMD oxidation. J. Mol. Cell Biol. 2019, 11, 1069–1082. [Google Scholar] [CrossRef] [Green Version]
  137. Abais, J.M.; Xia, M.; Zhang, Y.; Boini, K.M.; Li, P.L. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid. Redox Signal. 2015, 22, 1111–1129. [Google Scholar] [CrossRef] [Green Version]
  138. Martinon, F. Signaling by ROS drives inflammasome activation. Eur. J. Immunol. 2010, 40, 616–619. [Google Scholar] [CrossRef]
  139. Boaru, S.G.; Borkham-Kamphorst, E.; Van de Leur, E.; Lehnen, E.; Liedtke, C.; Weiskirchen, R. NLRP3 inflammasome expression is driven by NF-kB in cultured hepatocytes. Biochem. Biophys. Res. Commun. 2015, 458, 700–706. [Google Scholar] [CrossRef] [PubMed]
  140. Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 2010, 11, 136–140. [Google Scholar] [CrossRef] [PubMed]
  141. Ratajczak, M.Z.; Kucia, M. SARS-CoV-2 infection and overactivation of Nlrp3 inflammasome as a trigger of cytokine “storm” and risk factor for damage of hematopoietic stem cells. Leukemia 2020, 34, 1726–1729. [Google Scholar] [CrossRef]
  142. Donath, M.Y.; Böni-Schnetzler, M.; Ellingsgaard, H.; Halban, P.A.; Ehses, J.A. Cytokine production by islets in health and diabetes: Cellular origin, regulation and function. Trends Endocrinol. Metab. 2010, 21, 261–267. [Google Scholar] [CrossRef]
  143. Qiao, Y.; Wang, P.; Qi, J.; Zhang, L.; Gao, C. TLR-induced NF-kB activation regulates NLRP3 expression in murine macrophages. FEBS Lett. 2012, 586, 1022–1026. [Google Scholar] [CrossRef] [Green Version]
  144. Place, D.E.; Kanneganti, T.D. Recent advances in inflammasome biology. Curr. Opin. Immunol. 2018, 50, 32–38. [Google Scholar] [CrossRef]
  145. Zhao, M.; Bai, M.; Ding, G.; Zhang, Y.; Huang, S.; Jia, Z.; Zhang, A. Angiotensin II stimulates the NLRP3 inflammasome to induce podocyte injury and mitochondrial dysfunction. Kidney Dis. 2018, 4, 83–94. [Google Scholar] [CrossRef] [PubMed]
  146. Van den Berg, D.F.; Te Velde, A.A. Severe COVID-19: NLRP3 inflammasome dysregulated. Front. Immunol. 2020, 11, 1580. [Google Scholar] [CrossRef] [PubMed]
  147. De Flora, S.; Grassi, C.; Carati, L. Attenuation of influenza-like symptomatology and improvement of cell-mediated immunity with long-term N-acetylcysteine treatment. Eur. Respir. J. 1997, 10, 1535–1541. [Google Scholar] [CrossRef]
  148. McCarty, M.F.; DiNicolantonio, J.J. Nutraceuticals have potential for boosting the type 1 interferon response to RNA viruses including influenza and coronavirus. Prog. Cardiovasc. Dis. 2020, 63, 383–385. [Google Scholar] [CrossRef] [PubMed]
  149. De Flora, S.; Balansky, R.; La Maestra, S. Rationale for the use of N acetylcysteine in both prevention and adjuvant therapy of COVID-19. FASEB J. 2020, 34, 13185–13193. [Google Scholar] [CrossRef]
  150. Hati, S.; Bhattacharyya, S. Impact of thiol-disulfide balance on the binding of Covid-19 spike protein with angiotensin converting enzyme 2 receptor. CS Omega 2020, 5, 16292–16298. [Google Scholar] [CrossRef] [PubMed]
  151. Geiler, J.; Michaelis, M.; Naczk, P.; Leutz, A.; Langer, K.; Doerr, H.W.; Cinatl, J. N-acetyl-L-cysteine (NAC) inhibits virus replication and expression of pro-inflammatory molecules in A549 cells infected with highly pathogenic H5N1 influenza A virus. Bioch. Pharmacol. 2010, 79, 413–420. [Google Scholar] [CrossRef] [Green Version]
  152. Mata, M.; Morcillo, E.; Gimeno, C.; Cortijo, J. N-acetyl-L-cysteine (NAC) inhibit mucin syn-thesis and pro-inflammatory mediators in alveolar type II epithelial cells infected with influenza virus A and B and with respiratory syncytial virus (RSV). Bioch Pharmacol. 2011, 82, 548–555. [Google Scholar] [CrossRef] [Green Version]
  153. Bauer, S.R.; Kapoor, A.; Rath, M.; Thomas, S.A. What is the role of supplementation with ascorbic acid, zinc, vitamin D, or N-acetylcysteine for prevention or treatment of COVID-19? Cleve Clin. J. Med. 2020. [Google Scholar] [CrossRef]
  154. Šalamon, Š.; Kramar, B.; Marolt, T.P.; Poljšak, B.; Milisav, I. Medical and dietary uses of N-acetylcysteine. Antioxidants 2019, 8, 111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Guglielmetti, G.; Quaglia, M.; Sainaghi, P.P.; Castello, L.M.; Vaschetto, R.; Pirisi, M.; Corte, F.D.; Avanzi, G.C.; Stratta, P.; Cantaluppi, V. “War to the knife”against thromboinflammation to protect endothelial function of COVID-19 patients. Crit. Care. 2020, 24, 365. [Google Scholar] [CrossRef] [PubMed]
  156. Andreou, A.; Trantza, S.; Filippou, D.; Sipsas, N.; Tsiodras, S. COVID- 19: The potential role of copper and N-acetylcysteine [NAC] in a combination of candidate antiviral treatments against SARS-CoV-2. In Vivo 2020, 34, 1567–1588. [Google Scholar] [CrossRef]
  157. Jorge-Aarón, R.M.; Rosa-Ester, M.P. N-acetylcysteine as a potential treatment for novel coronavirus disease 2019 [published online ahead of print, 2020 Jul 14]. Future Microbiol. 2020, 5, 959–962. [Google Scholar] [CrossRef] [PubMed]
  158. Poe, F.L.; Corn, J. N-Acetylcysteine: A potential therapeutic agent for SARS-CoV-2. Med. Hypotheses 2020, 143, 109862. [Google Scholar] [CrossRef]
  159. Memorial Sloan Kettering Cancer Center. A Study of N-Acetylcysteine in Patients with COVID-19 Infection 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT04374461 (accessed on 31 December 2020).
  160. Kensler, T.W.; Wakabayashi, N.; Biswal, S. Cell survival responses to environmental stresses via the Keap1-NRF2-ARE pathway. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 89–116. [Google Scholar] [CrossRef]
  161. Sadowska, A.M. N-Acetylcysteine mucolysis in the management of chronic obstructive pulmonary disease. Ther. Adv. Respir. Dis. 2012, 6, 127–135. [Google Scholar] [CrossRef] [Green Version]
  162. Hui, D.S.C.; Lee, N. Adjunctive therapies and immunomodulating agents for severe influenza. Influenza Other Respir. Viruses 2013, 7, 52–59. [Google Scholar] [CrossRef]
  163. Crinelli, R.; Zara, C.; Smietana, M.; Retini, M.; Magnani, M.; Fraternale, A. Boosting GSH using the Co-drug approach: I-152, a conjugate of N-acetyl-cysteine and mercaptoethylamine. Nutrients 2019, 11, 1291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Horowitz, R.I.; Freeman, P.M.; Bruzzese, J. Efficacy of gluthahione therapy in relieving dyspnea associated with COVID-19 pneumonia. A report of 2 cases. Respir. Med. Case Rep. 2020, 30, 101063. [Google Scholar]
  165. Marik, P.E. Vitamin C for the treatment of sepsis: The scientific rationale. Pharmacol. Ther. 2018, 189, 63–70. [Google Scholar] [CrossRef]
  166. Mohammed, B.M.; Fisher, B.J.; Kraskauskas, D.; Farkas, D.; Brophy, D.F.; Fowler, A.A., III; Natarajan, R. Vitamin C: A novel regulator of neutrophil extracellular trap formation. Nutrients 2013, 5, 3131–3151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Fuchs, T.A.; Abed, U.; Goosmann, C.; Hurwitz, R.; Schulze, I.; Wahn, V. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 2007, 176, 231–241. [Google Scholar] [CrossRef] [PubMed]
  168. Radermecker, C.; Detrembleur, N.; Guiot, J.; Cavalier, E.; Henket, M. Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. J Exp Med. 2020, 217, e20201012. [Google Scholar] [CrossRef] [PubMed]
  169. Abobaker, A.; Alzwi, A.; Alraied, A.H.A. Overview of the possible role of vitamin C in management of COVID-19. Pharmacol. Rep. 2020, 72, 1517–1528. [Google Scholar] [CrossRef]
  170. Marik, P.E.; Kory, P.; Varon, J.; Iglesias, J.; Meduri, G.U. MATH+ protocol for the treatment of SARS-CoV-2 infection: The scientific rationale. Expert Rev. Anti Infect. Ther. 2020, 10, 1–7. [Google Scholar] [CrossRef] [PubMed]
  171. Domain-Specific Appendix: VITAMIN, C. REMAP-CAP: Randomized, Embedded, Multifactorial Adaptive Platform Trial for Community-Acquired Pneumonia 2020. Available online: https://static1.squarespace.com/static/5cde3c7d9a69340001d79e/t/5f1bba732cda7f10310643fe/1595652735252/REMAP-CAP+Vitamin+C+Domain+Specific+Appendix+V2+-08+June+2020_WM.pdf (accessed on 31 December 2020).
  172. Hiedra, R.; Lo, K.B.; Elbashabsheh, M.; Gul, F.; Wright, R.M.; Albano, J.; Azmaiparashvili, Z.; Patarroyo Aponte, G. The use of IV vitamin C for patients with COVID-19: A case series. Expert Rev. Anti Infect. Ther. 2020, 18, 1259–1261. [Google Scholar] [CrossRef]
  173. Komaravelli, N.; Casola, A. Respiratory Viral Infections and Subversion of Cellular Antioxidant Defenses. J. Pharm. Pharm. 2014, 5, 1000141. [Google Scholar]
  174. Wyler, E.; Franke, V.; Menegatti, J.; Kocks, C.; Boltengagen, A.; Praktiknjo, S.; Walch-Rückheim, B.; Bosse, J.; Rajewsky, N.; Grässer, F.; et al. Single-cell RNA-sequencing of herpes simplexvirus 1-infected cells connects NRF2 activation to an antiviral program. Nat. Commun. 2019, 10, 4878. [Google Scholar] [CrossRef] [Green Version]
  175. Halder, U.C.; Bagchi, P.; Chattopadhyay, S.; Dutta, D.; Chawla-Sarkar, M. Cell death regulation during influenza A virus infection by matrix [M1] protein: A model of viral control over the cellular survival pathway. Cell Death Dis. 2011, 2, e197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Olagnier, D.; Farahani, E.; Thyrsted, J.; Cadanet, J.B.; Herengt, A.; Idorn, M.; Hait, A.; Hernaez, B.; Knudsen, A.; Iversen, M.B.; et al. Identification of SARSCoV2-mediated suppression of NRF2 signaling reveals a potent antiviral and anti-inflammatory activity of 4-octyl-itaconate and dimethyl fumarate. Nat. Commun. 2020, 11, 4938. [Google Scholar] [CrossRef]
  177. Cuadrado, A.; Martín-Moldes, Z.; Ye, J.; Lastres-Becker, I. Transcription factors NRF2 and NF-kappaB are coordinated effectors of the Rho family, GTP binding protein RAC1 during inflammation. J. Biol. Chem. 2014, 289, 15244–15258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Lastres-Becker, I.; Innamorato, N.G.; Jaworski, T.; Rábano, A.; Kügler, S.; Van Leuven, F.; Cuadrado, A. Fractalkine activates NRF2/NFE2L2 and heme oxygenase 1 to restrain tauopathy induced microgliosis. Brain 2014, 137, 78–91. [Google Scholar] [CrossRef] [PubMed]
  179. Boutten, A.; Goven, D.; Artaud-Macari, E.; Boczkowski, J.; Bonay, M. NRF2 targeting: A promising therapeutic strategy in chronic obstructive pulmonary disease. Trends Mol. Med. 2011, 17, 363–371. [Google Scholar] [CrossRef]
  180. DeDiego, M.L.; Nieto-Torres, J.L.; Regla-Nava, J.A.; Jimenez-Guardeño, J.M.; Fernandez-Delgado, R.; Fett, C.; Castaño-Rodriguez, C.; Perlman, S.; Enjuanes, L. Inhibition of NF-kappaB-mediated inflammation in severe acute respiratory syndrome coronavirus-infected mice increases survival. J. Virol. 2014, 88, 913–924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Dosch, S.F.; Mahajan, S.D.; Collins, A.R. SARS coronavirus spike protein-induced innate immune response occurs via activation of the NF-kappaB pathway in human monocyte macrophages in vitro. Virus Res. 2009, 142, 19–27. [Google Scholar] [CrossRef]
  182. Cuadrado, A.; Pajares, M.; Benito, C.; Jiménez-Villegas, J.; Escoll, M.; Fernández-Ginés, R.; Garcia Yagüe, A.J.; Lastra, D.; Manda, G.; Ana, I.; et al. Can activation of NRF2 be a strategy against COVID-19? Trends Pharmacol. Sci. 2020, 41, 598–610. [Google Scholar] [CrossRef]
  183. Espinoza, J.A.; González, P.A.; Kalergis, A.M. Modulation of antiviral immunity by heme oxygenase-1. Am. J. Pathol. 2017, 187, 487–493. [Google Scholar] [CrossRef] [Green Version]
  184. Wagener, F.A.D.T.G.; Pickkers, P.; Peterson, S.J.; Immenschuh, S.; Abraham, N.G. Targeting the heme-heme oxygenase system to prevent severe complications following COVID-19 infections. Antioxidants 2020, 9, 540. [Google Scholar] [CrossRef]
  185. McCord, J.M.; Hybertson, B.M.; Cota-Gomez, A.; Geraci, K.P.; Gao, B. NRF2 Activator PB125 as a Potential Therapeutic Agent against COVID-19. Antioxidants 2020, 9, 518. [Google Scholar] [CrossRef] [PubMed]
  186. Dittmann, M.; Hoffmann, H.H.; Bieniasz, P.D.; Rice, C.M. A Serpin Shapes the Extracellular Environment to Prevent Influenza A Virus Maturation. Cell 2015, 160, 631–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Mrityunjaya, M.; Pavithra, V.; Neelam, R.; Janhavi, P.; Halami, P.M.; Ravindra, P.V. Immune-Boosting, Antioxidant and Anti-inflammatory Food Supplements Targeting Pathogenesis of COVID-19. Front. Immunol. 2020, 11, 570122. [Google Scholar] [CrossRef] [PubMed]
  188. Lucas, J.M.; Heinlein, C.; Kim, T.; Hernandez, S.A.; Malik, M.S.; True, L.D.; Morrissey, C.; Corey, E.; Montgomery, B.; Mostaghel, E.; et al. The androgen-regulated protease TMPRSS2 activates a proteolytic cascade involving components of the tumor microenvironment and promotes prostate cancer metastasis. Cancer Discov. 2014, 4, 1310–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Zhang, S.; Jiang, J.; Ren, Q.; Jia, Y.; Shen, J.; Shen, H.; Lin, X.; Lu, H.; Xie, Q. Ambroxol inhalation ameliorates LPS-induced airway inflammation and mucus secretion through the extracellular signal-regulated kinase 1/2 signaling pathway. Eur. J. Pharmacol. 2016, 775, 138–148. [Google Scholar] [CrossRef] [PubMed]
  190. Sunkari, S.; Thatikonda, S.; Pooladanda, V.; Challa, V.S.; Godugu, C. Protective effects of ambroxol in psoriasis like skin inflammation: Exploration of possible mechanisms. Intern. Immunopharmacol. 2019, 71, 301–312. [Google Scholar] [CrossRef] [PubMed]
  191. Ansarin, K.; Tolouian, R.; Ardalan, M.; Taghizadieh, A.; Varshochi, M.; Teimouri, S.; Vaezi, T.; Valizadeh, H.; Saleh, P.; Safiri, S.; et al. Effect of bromhexine on clinical outcomes and mortality in COVID-19 patients: A randomized clinical trial. BioImpacts 2020, 10, 209–215. [Google Scholar] [CrossRef]
  192. Montes Diaz, G.; Hupperts, R.; Fraussen, J.; Somers, V. Dimethyl fumarate treatment in multiple sclerosis: Recent advances in clinical and immunological studies. Autoimmun. Rev. 2018, 17, 1240–1250. [Google Scholar] [CrossRef]
  193. Meng, Y.; Li, T.; Zhou, G.S.; Chen, Y.; Yu, C.H.; Pang, M.X.; Li, W.; Li, Y.; Zhang, W.Y.; Li, X. The angiotensin-converting enzyme 2/angiotensin [1–7]/mas axis protects against lung fibroblast migration and lung fibrosis by inhibiting the NOX4-derived ROS-mediated RhoA/Rho kinase pathway. Antioxid. Redox. Signal. 2015, 22, 241–258. [Google Scholar] [CrossRef]
  194. Lei, C.; Qian, K.; Li, T.; Zhang, S.; Fu, W.; Ding, M.; Hu, S. Neutralization of SARS-CoV-2 spike pseudotyped virus by recombinant ACE2-Ig. Nat. Commun. 2020, 11, 2070. [Google Scholar] [CrossRef] [Green Version]
  195. Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.A.; Stahl, M.; Leopoldi, A.; Garreta, E.; Hurtado Del Pozo, C.; Prosper, F.; et al. Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell 2020, 181, 905–913. [Google Scholar] [CrossRef] [PubMed]
  196. Zoufaly, A.; Poglitsch, M.; Aberle, J.H.; Hoepler, W.; Seitz, T.; Traugott, M.; Grieb, A.; Pawelka, E.; Laferl, H.; Wenisch, C.; et al. Human recombinant soluble ACE2 in severe COVID-19. Lancet Respir. Med. 2020, 8, 1154–1158. [Google Scholar] [CrossRef]
  197. Shah, A. Novel Coronavirus-Induced NLRP3 Inflammasome Activation: A Potential Drug Target in the Treatment of COVID-19. Front. Immunol. 2020, 11, 1021. [Google Scholar] [CrossRef]
  198. He, H.; Jiang, H.; Chen, Y.; Ye, J.; Wang, A.; Wang, C.; Liu, Q.; Liang, G.; Deng, X.; Jiang, W.; et al. Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity. Nat. Commun. 2018, 9, 2550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Juliana, C.; Fernandes-Alnemri, T.; Wu, J.; Datta, P.; Solorzano, L.; Yu, J.W.; Meng, R.; Quong, A.A.; Latz, E.; Scott, C.P.; et al. Anti-inflammatory compounds parthenolide and bay 11-7082 are direct inhibitors of the inflammasome. J. Biol. Chem. 2010, 285, 9792–9802. [Google Scholar] [CrossRef] [Green Version]
  200. Costa, L.S.; Outlioua, A.; Anginot, A.; Akarid, K.; Arnoult, D. RNA viruses promote activation of the NLRP3 inflammasome through cytopathogenic effect-induced potassium efflux. Cell Death Dis. 2019, 10, 346. [Google Scholar] [CrossRef]
  201. Lamkanfi, M.; Mueller, J.L.; Vitari, A.C.; Misaghi, S.; Fedorova, A.; Deshayes, K.; Lee, W.P.; Hoffman, H.M.; Dixit, V.M. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. J. Cell Biol. 2009, 187, 61–70. [Google Scholar] [CrossRef] [Green Version]
  202. Huang, Y.; Jiang, H.; Chen, Y.; Wang, X.; Yang, Y.; Tao, J.; Deng, X.; Liang, G.; Zhang, H.; Jiang, W.; et al. Tranilast directly targets NLRP 3 to treat inflammasome-driven diseases. EMBO Mol. Med. 2018, 10, e8689. [Google Scholar] [CrossRef]
  203. Leung, Y.Y.; Yao Hui, L.L.; Kraus, V.B. Colchicine—update on mechanisms of action and therapeutic uses. Semin. Arthritis Rheum. 2015, 45, 341–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Misawa, T.; Takahama, M.; Kozaki, T.; Lee, H.; Zou, J.; Saitoh, T.; Akira, S. Microtubule driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat. Immunol. 2013, 14, 454–460. [Google Scholar] [CrossRef]
  205. Daniels, M.J.; Rivers-Auty, J.; Schilling, T.; Spencer, N.G.; Watremez, W.; Fasolino, V.; Booth, S.J.; White, C.S.; Baldwin, A.G.; Freeman, S.; et al. Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models. Nat Commun. 2016, 7, 12504. [Google Scholar] [CrossRef] [Green Version]
  206. Rothan, H.A.; Bahrani, H.; Abdulrahman, A.Y.; Mohamed, Z.; Teoh, T.C.; Othman, S.; Rashid, N.N.; Rahman, N.A.; Yusof, R. Mefenamic acid in combination with ribavirin shows significant effects in reducing chikungunya virus infection in vitro and in vivo. Antiviral. Res. 2016, 127, 50–56. [Google Scholar] [CrossRef]
  207. Cain, D.W.; Cidlowski, J.A. Immune regulation by glucocorticoids. Nat Rev. Immunol 2017, 17, 233–247. [Google Scholar] [CrossRef] [PubMed]
  208. Busillo, J.M.; Cidlowski, J.A. The five Rs of glucocorticoid action during inflammation: Ready, reinforce, repress, resolve, and restore. Trends Endocrinol. Metab. 2013, 24, 109–119. [Google Scholar] [CrossRef] [Green Version]
  209. Quatrini, L.; Ugolini, S. New insights into the cell- and tissue-specificity of glucocorticoid actions. Cell. Mol. Immunol. 2020, 18, 1–10. [Google Scholar] [CrossRef] [PubMed]
  210. Yang, N.; Zhang, W.; Shi, X.M. Glucocorticoid-induced leucine zipper (GILZ) mediates glucocorticoid action and inhibits inflammatory cytokine-induced COX-2 expression. J. Cell. Biochem. 2008, 103, 1760–1771. [Google Scholar] [CrossRef] [PubMed]
  211. Mitroulis, I.; Alexaki, V.I.; Kourtzelis, I.; Ziogas, A.; Hajishengallis, G.; Chavakis, T. Leukocyte integrins: Role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol. Ther. 2015, 147, 123–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Zielinska, K.A.; Van Moortel, L.; Opdenakker, G.; De Bosscher, K.; Van den Steen, P.E. Endothelial response to glucocorticoids in inflammatory diseases. Front Immunol 2016, 7, 592. [Google Scholar] [CrossRef]
  213. Busillo, J.M.; Ehrchen, J.; Steinmuller, L.; Barczyk, K. Glucocorticoids induce differentiation of a specifically activated, anti-inflammatory subtype of human monocytes. Blood 2007, 109, 1265–1274. [Google Scholar]
  214. Meers, G.K.; Bohnenberger, H.; Reichardt, H.M.; Luhder, F.; Reichardt, S.T. Impaired resolution of DSS-induced colitis in mice lacking the glucocorticoid receptor inmyeloid cells. PLoS ONE 2018, 13, e0190846. [Google Scholar] [CrossRef] [PubMed]
  215. Vallelian, F.; Schaer, C.A.; Kaempfer, T.; Ghering, P.; Duerst, E.; Schoedon, G.; Schaer, D.J. Glucocorticoid treatment skews human monocyte differentiation into a hemoglobin-clearance phenotype with enhanced heme-iron recycling and antioxidant capacity. Blood 2010, 116, 5347–5356. [Google Scholar] [CrossRef] [Green Version]
  216. RECOVERY Collaborative Group; Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; et al. Dexamethasone in Hospitalized Patients with Covid-19 - Preliminary Report. N. Engl. J. Med. 2020. [Google Scholar] [CrossRef]
  217. The WHO Rapid Evidence Appraisal for COVID-19 Therapies (REACT) Working Group. Association between administration of systemic corticosteroids and mortality among critically ill Patients with COVID-19. A Meta-analysis. JAMA 2020, 324, 1330–1341. [Google Scholar]
  218. Vane, J.R.; Botting, R.M. Mechanism of action of nonsteroidal anti-inflammatory drugs. Am. J. Med. 1998, 104, 2S–8S. [Google Scholar] [CrossRef]
  219. Day, M. Covid-19: Ibuprofen should not be used for managing symptoms, say doctors and scientists. BMJ 2020, 368, m1086. [Google Scholar] [CrossRef] [Green Version]
  220. FitzGerald, G.A. Misguided drug advice for COVID-19. Science 2020, 367, 1434. [Google Scholar] [PubMed] [Green Version]
  221. Little, P. Non-steroidal anti-inflammatory drugs and covid-19. BMJ 2020, 368, m1185. [Google Scholar] [CrossRef] [Green Version]
  222. Huh, K.; Ji, W.; Kang, M.; Hong, J.; Bae, G.H.; Lee, R.; Na, Y.; Jung, J. Association of prescribed medications with the risk of COVID-19 infection and severity among adults in South Korea. Int. J. Infect. Dis. 2020, 104, 7–14. [Google Scholar] [CrossRef]
  223. Freites Nuñez, D.D.; Leon, L.; Mucientes, A.; Rodriguez-Rodriguez, L.; Font Urgelles, J.; Madrid García, A.; Colomer, J.I.; Jover, J.A.; Fernandez-Gutierrez, B.; Abasolo, L. Risk factors for hospital admissions related to COVID-19 in patients with autoimmune inflammatory rheumatic diseases. Ann. Rheum. Dis. 2020, 79, 1393–1399. [Google Scholar] [CrossRef]
  224. Imam, Z.; Odish, F.; Gill, I.; O’Connor, D.; Armstrong, J.; Vanood, A.; Ibironke, O.; Hanna, A.; Ranski, A.; Halalau, A. Older age and comorbidity are independent mortality predictors in a large cohort of 1305 COVID-19 patients in Michigan, United States. J. Intern. Med. 2020, 288, 469–476. [Google Scholar] [CrossRef]
  225. Hong, W.; Chen, Y.; You, K.; Tan, S.; Wu, F.; Tao, J.; Chen, X.; Zhang, J.; Xiong, Y.; Yuan, F. Celebrex Adjuvant Therapy on Coronavirus Disease 2019: An Experimental Study. Front Pharmacol. 2020, 11, 561674. [Google Scholar] [CrossRef]
  226. EMA advice on the use of NSAIDs for Covid-19. Drug Ther. Bull. 2020, 58, 69. [CrossRef] [PubMed]
  227. Robb, C.T.; Goepp, M.; Rossi, A.G.; Yao, C. Non-steroidal anti-inflammatory drugs, prostaglandins, and COVID-19. Br. J Pharmacol. 2020, 177, 4899–4920. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (a) SARS-CoV-2 structure; (b) Schematic drawing of SARS-CoV-2 Spike (S) protein. S1, receptor-binding subunit; S2, membrane fusion subunit; FP, fusion protein; RBD, receptor binding domain.
Figure 1. (a) SARS-CoV-2 structure; (b) Schematic drawing of SARS-CoV-2 Spike (S) protein. S1, receptor-binding subunit; S2, membrane fusion subunit; FP, fusion protein; RBD, receptor binding domain.
Antioxidants 10 00272 g001
Figure 2. Schematic diagram of SARS-CoV-2 effects on renin angiotensin system. ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; Ang II, Angiotensin II; Adam-17, a disintegrin and metalloproteinase17; AT1R, angiotensin II type-1 receptor; TMPRSS2, transmembrane protease serine 2.
Figure 2. Schematic diagram of SARS-CoV-2 effects on renin angiotensin system. ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; Ang II, Angiotensin II; Adam-17, a disintegrin and metalloproteinase17; AT1R, angiotensin II type-1 receptor; TMPRSS2, transmembrane protease serine 2.
Antioxidants 10 00272 g002
Figure 3. Oxidative stress and inflammation induced by SARS-CoV-2 infection. ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; AT1R, angiotensin II type-1 receptor; NOX2, NADPH oxidase 2, NF-kB, nuclear factor kB; ROS, reactive oxygen species; TRXIP, thioredoxin interacting/inhibiting protein; NLRP3, NOD-like receptor protein 3.
Figure 3. Oxidative stress and inflammation induced by SARS-CoV-2 infection. ACE, angiotensin-converting enzyme; ACE2, angiotensin-converting enzyme 2; AT1R, angiotensin II type-1 receptor; NOX2, NADPH oxidase 2, NF-kB, nuclear factor kB; ROS, reactive oxygen species; TRXIP, thioredoxin interacting/inhibiting protein; NLRP3, NOD-like receptor protein 3.
Antioxidants 10 00272 g003
Figure 4. Potential beneficial effects of Nrf2 activators against SARS-CoV2-infection. NRF2, nuclear factor erythroid 2 p45-related factor 2; NF-kB, nuclear Factor kB; TMPRSS2, transmembrane protease serine 2.
Figure 4. Potential beneficial effects of Nrf2 activators against SARS-CoV2-infection. NRF2, nuclear factor erythroid 2 p45-related factor 2; NF-kB, nuclear Factor kB; TMPRSS2, transmembrane protease serine 2.
Antioxidants 10 00272 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fratta Pasini, A.M.; Stranieri, C.; Cominacini, L.; Mozzini, C. Potential Role of Antioxidant and Anti-Inflammatory Therapies to Prevent Severe SARS-Cov-2 Complications. Antioxidants 2021, 10, 272. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10020272

AMA Style

Fratta Pasini AM, Stranieri C, Cominacini L, Mozzini C. Potential Role of Antioxidant and Anti-Inflammatory Therapies to Prevent Severe SARS-Cov-2 Complications. Antioxidants. 2021; 10(2):272. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10020272

Chicago/Turabian Style

Fratta Pasini, Anna M., Chiara Stranieri, Luciano Cominacini, and Chiara Mozzini. 2021. "Potential Role of Antioxidant and Anti-Inflammatory Therapies to Prevent Severe SARS-Cov-2 Complications" Antioxidants 10, no. 2: 272. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox10020272

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