Next Article in Journal
Beneficial Effects of Opuntia humifusa (Korean Cheonnyuncho) on Human Health Based on Antioxidant Properties: Systematic Review and Meta-Analysis
Next Article in Special Issue
Novel Hydrogen Sulfide Hybrid Derivatives of Keap1-Nrf2 Protein–Protein Interaction Inhibitor Alleviate Inflammation and Oxidative Stress in Acute Experimental Colitis
Previous Article in Journal
Engineered Adipose-Derived Stem Cells Overexpressing RXFP1 via CRISPR Activation Ameliorate Erectile Dysfunction in Diabetic Rats
Previous Article in Special Issue
Maternal Calorie Restriction Induces a Transcriptional Cytoprotective Response in Embryonic Liver Partially Dependent on Nrf2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Is Nuclear Factor Erythroid 2-Related Factor 2 a Target for the Intervention of Cytokine Storms?

1
The First Department of Clinical Medicine, China Medical University, Shenyang 110122, China
2
Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang 110122, China
3
Group of Chronic Disease and Environmental Genomics, School of Public Health, China Medical University, Shenyang 110122, China
4
Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2023, 12(1), 172; https://doi.org/10.3390/antiox12010172
Submission received: 10 December 2022 / Revised: 8 January 2023 / Accepted: 9 January 2023 / Published: 11 January 2023

Abstract

:
The term “cytokine storm” describes an acute pathophysiologic state of the immune system characterized by a burst of cytokine release, systemic inflammatory response, and multiple organ failure, which are crucial determinants of many disease outcomes. In light of the complexity of cytokine storms, specific strategies are needed to prevent and alleviate their occurrence and deterioration. Nuclear factor erythroid 2-related factor 2 (NRF2) is a CNC-basic region-leucine zipper protein that serves as a master transcription factor in maintaining cellular redox homeostasis by orchestrating the expression of many antioxidant and phase II detoxification enzymes. Given that inflammatory response is intertwined with oxidative stress, it is reasonable to assume that NRF2 activation limits inflammation and thus cytokine storms. As NRF2 can mitigate inflammation at many levels, it has emerged as a potential target to prevent and treat cytokine storms. In this review, we summarized the cytokine storms caused by different etiologies and the rationale of interventions, focusing mainly on NRF2 as a potential therapeutic target.

1. Introduction

The cytokine storm was first described in medical literature in 1993 and recognized later as a series of exaggerated immune responses characterized by a burst of pro-inflammatory cytokine release, which usually causes acute systemic inflammatory response [1]. While there is still no widely accepted definition for cytokine storm, three key characteristics, including elevated circulating cytokines, acute systemic inflammation, and secondary multi-organ dysfunction, have been proposed as the identification criteria [2]. A cytokine storm is generally divided into the following three stages: primary loci stage, systemic inflammation, and multi-organ failure [2]. Although multiple general recommendations on anti-inflammatory treatments have been proposed for cytokine storms associated with a variety of diseases, including malignancy, rheumatologic disease, sepsis syndrome, and severe coronavirus disease 2019 (COVID-19) [2,3,4], there is still an urgent need to find novel strategies and methods to prevent and alleviate cytokine storms.
Nuclear factor erythroid 2-related factor 2 (NRF2) is a CNC-basic region-leucine zipper (CNC-bZIP) protein that functions as a key transcription factor regulating the cellular response against oxidative and electrophilic stresses. Kelch-like ECH-associated protein 1 (KEAP1), a substrate adaptor for the Cullin 3 (Cul3)-based E3 ubiquitin ligase complex, acts as a stress sensor and negative regulator for NRF2 stability [5]. Under unstressed conditions, KEAP1 sequesters NRF2 and mediates its degradation, which maintains NRF2 at low levels [6]. In response to oxidative and/or electrophilic challenge, the key cysteine residues on the KEAP1 could be oxidized, leading to conformation change and subsequently losing its function as a substrate adaptor for ubiquitin ligase, facilitating the escape of NRF2 from ubiquitination and nuclear accumulation. In the nucleus, NRF2 may form a heterodimer with small musculoaponeurotic fibrosarcoma (sMaf) proteins and bind to the antioxidant response elements (AREs), resulting in the transcription of a battery of target genes, which encode antioxidants, phase II detoxification enzymes, drug transporters and key molecules controlling cell metabolism [7,8,9]. In addition, accumulating evidence has shown that activation of the NRF2-ARE signaling pathway is crucial to the alleviation of inflammation. On the one hand, NRF2-induced cytoprotective proteins are likely to be important in the regulation of innate immune responses by repressing the expression of pro-inflammatory genes and potentiating the anti-inflammatory signaling [10]. On the other hand, NRF2-mediated antioxidant response might mitigate the activation of nuclear factor kappa-light chain-enhancer of activated B (NF-κB) to reduce the inflammatory response [11]. The absence of NRF2 exacerbates NF-κB activity, leading to increased cytokine production, whereas NF-κB, in turn, can modulate NRF2 transcription and activity [12]. In addition, NRF2 has also been found to regulate the transcription of a variety of cytokines in distinct ways.
Although the anti-inflammatory effects of NRF2 have been mainly attributed to its regulation in antioxidative genes, the molecular details underlying the effects are only partially characterized [10,13,14,15]. In light of the successful applications of multiple NRF2 activators in treating inflammation-related disorders, the effects of NRF2 activation against cytokine storms have been actively investigated. The present review provides an overview of the cytokine storms caused by different etiologies and the rationale of interventions, focusing particularly on NRF2 as a potential therapeutic target.

2. Cytokine Storms

Cytokine storms result from the collapse of complex immune networks, of which the key point is the disparity in forces between the pro-inflammatory and anti-inflammatory systems [2,16,17]. Cytokine storms can be divided into the following four types according to their initiating factors: pathogen, clinical treatment, autoimmune, and others [17,18,19,20]. In the case of pathogen-induced cytokine storms, disseminated bacteria or virus infections inducing sepsis and abnormally mobilized host immune system are evident [21,22]. A fraction of COVID-19 patients suffered from a life-threatening systemic inflammatory state, which was first described in children and adolescents, was termed as multisystem inflammatory syndrome [23]. In addition, a similar condition in adults has also been reported as multisystem inflammatory syndrome [24]. In COVID-19 patients, plasma levels of pro-inflammatory cytokines, including interleukin (IL)-1α, IL-1β, and tumor necrosis factor α (TNF-α), are elevated, compared to healthy adults [25,26]. In line with the findings in COVID-19 patients, induction of type I and III interferons (IFN), which are considered as the first line of cellular antiviral defenses, was observed to be delayed in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), resulting in a subsequent increase of IFN-γ and TNF-α, which trigger inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 [21,27]. In addition, sepsis patients also show higher plasma levels of pro-inflammatory cytokines, including IL-6, IFN-γ, and TNF-α [28]. In contrast to pathogen-induced cytokine storms, treatment-induced cytokine storms are increasingly familiar to clinicians as more immune-cell engaging immunotherapies are used in clinics. For example, the cytokines released by chimeric antigen receptor (CAR) T-cells or macrophages activated in CAR T-cell therapy are the initiators of treatment-induced cytokine storms [29]. In the patients with CAR T-cell therapy, the serum levels of pro-inflammatory cytokines, including IL-6 and IFN-γ, elevated with the onset of cytokine storms [30]. In agreement with the effects of CAR T-cell therapy, cytokine storms could also happen in some patients with autoimmune disorders, such as familial Mediterranean fever and hemophagocytic lymphohistiocytosis. In these patients, innate cells abnormally release TNF-α, IFN-γ, IL-1β, and IL-6, and trigger cytokine storms [2,31,32,33]. Some non-infectious diseases, such as acute pancreatitis, may also induce cytokine storms [34]. These non-infectious diseases are not caused by specific pathogens but by some physical or chemical factors, which could trigger a series of excessive immune responses, leading to cytokine storms. In acute pancreatitis patients, high serum or plasma levels of IL-6 and IL-8 were observed in severe versus non-severe cases [35]. In general, cytokine storms are associated with immune disorders when the immune system of the host is mobilized toward a higher level. In contrast, regulatory T-cells (Tregs) can reduce the primary inflammatory responses and the tissue damage caused by systemic inflammation [36,37]. It is worth noting that the increase in the negative regulatory factors of inflammatory responses might be related to a period of low immune response called immunoparalysis, which increases the risk of sepsis and organ failure after a cytokine storm and leads to a poor prognosis [36,38].
In pathogen-induced cytokine storms, as illustrated in Figure 1, the pathogen-associated molecular patterns (PAMPs) derived from various microorganisms, such as influenza A virus (IAV), SARS-CoV-2, and Staphylococcus aureus, might induce abnormal activation of primary inflammatory responses at the affected loci. The PAMPs that induce cytokine storms include RNA from viruses and superantigens from bacteria [39,40,41]. In the process, the characteristics of the PAMPs are crucial [42,43], as the infected cells may release a variety of damage-associated molecular patterns (DAMPs) in a PAMPs-dependent manner. In the case of virus infection, pattern recognition receptors (PRRs) sense PAMPs, which are often aberrant RNA structures formed during virus replication [44]. For example, the RNA of SARS-CoV-19 can stimulate membrane-bound PRR, Toll-like receptor (TLR)-3, 7, and cytosolic PRR, melanoma differentiation-associated gene 5, and lead to the release of cytokines, including IL-6 and TNF [39]. The DAMPs, such as reactive oxygen species (ROS) and cytokines, may form a positive feedback loop to elicit the primary acute inflammatory effects, leading to a pathological burst of pro-inflammatory cytokines with mismatched secretion of anti-inflammatory cytokines into the systemic circulation [45].
The primary acute inflammatory responses at the infection loci may develop into systemic inflammatory outcomes when ROS, cytokines, and other DAMPs are poured into the systemic circulation and spread throughout the body [16,17,46]. The systemic inflammatory effects, commonly observed in patients with cytokine storms, are fever, coagulopathies, changes in the fibrinolysis system and vascular tension, endothelial dysfunction, and anasarca. These changes are generally associated with increased levels of TNF-α, IFN-γ, IL-1β, and IL-6 in the systemic circulation [2,47]. In addition, the systemic inflammatory effects cause secondary damage to the primary lesion loci and distant tissues, including the vascular and lymphatic systems, central or peripheral nervous system, gastrointestinal system, lung, heart, liver, kidney, and skin. While TNF-α, IL-1β, and IL-6 are the most important cytokines in this damaging process [21,48,49,50], in the later phase of critical infectious disorders, such as severe COVID-19 patients, the systemic levels of IL-4, IL-10, and IL-13 may also increase [16,38].
The key cellular events in cytokine storms, including cell proliferation, polarization, differentiation, maturation, apoptosis, and cytokine release, are modulated by multiple signaling pathways. For example, in pathogen-induced cytokine storms, PAMP-induced DAMPs are recognized by PRRs (e.g., TLRs or nucleotide-binding oligomerization domain-containing protein 2 (NOD2)) that trigger pro-inflammatory signaling. It has been well documented that mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) and NF-κB pathways are commonly involved in inflammatory processes [51,52,53]. These pathways lead the primary immune cells toward inflammatory responses and control the expression of cytokines and other inflammatory mediators. As a result, the activated primary immune cells release aberrantly high levels of cytokines into the systemic circulation. For instance, virus replication is normally first detected by PRRs and induces transcriptional activation of IFN-III and IFN-I mediated by NF-κB and interferon regulator factors, respectively [54,55]. Next, upregulation of IFN-stimulated genes and recruitment and coordination of specific subsets of leukocytes is triggered. Interestingly, when SARS-CoV-2 first replicates at a low level, the IFN-dependent pathways are suppressed, which could lead to antivirus defense failure. In contrast, high titers of SARS-CoV-2 induce a strong IFN-dependent response, elevated chemokines and high expression of IL-6 [27]. In addition, Janus kinase-signal transducer and activators of transcription (JAK-STAT), MAPK-ERK, and NF-κB pathways are also involved in the downstream signal transduction of cytokines in target cells [56,57].
In summary, cytokine storm is a state of auto-amplifying cytokine production due to dysregulated host immune response to different triggers, including severe infections, diseases and immunotherapies. The excessive activation of immune cells and uncontrolled generation of pro-inflammatory cytokines may further result in systemic inflammation across multiple systems, leading to organ dysfunction and failure if not treated adequately.

3. Crosstalk between Oxidative Stress and Inflammatory Response

Oxidative stress is commonly described as a persistent imbalance between ROS production and its breakdown by distinct antioxidant systems in favor of oxidation, leading to disruption of redox signaling and/or oxidative damage [58,59,60]. In general, overproduced superoxide anion (O2•−) and nitric oxide (NO) and their derivatives, including hydrogen peroxide (H2O2), hydroxyl radical (HO•−), and peroxynitrite (ONOO•−) generated from pathophysiologic processes and subsequent free radical chain reactions, are the primary initiating factors to induce oxidative stress.
ROS can be produced at many sites via multiple enzymes, including mitochondria, NADPH oxidases (NOX), and NO synthetases [61,62,63,64]. While ROS are crucial to eliminate pathogens, they may also cause damage to local tissues and trigger various immune events. In infection-induced cytokine storms, the PAMPs derived from pathogens might cause damage to parenchymal cells via PRRs or even induce regulated cell death (RCD), which may generate and release more DAMPs [65,66,67]. ROS, on the one hand, may stimulate the expression and secretion of many cytokines, such as IL-1β, TNF-α, and IL-6. In turn, increased cytokines promote the activation of NOX, leading to more ROS production, forming a vicious cycle; on the other hand, ROS could also function as a signaling molecule to initiate the activation of NRF2-mediated antioxidant response and thus stop the ROS-cytokine loop. To understand the details involving the paradoxical roles of ROS in inflammatory response, we conducted a series of computational modeling studies. Our mathematical models showed that the concentration effect of NRF2 activation induced by ROS, such as H2O2, displays a nonlinear relationship [8,68]. When H2O2 is at physiologically relevant low levels, it cannot induce oxidation of KEAP1 and subsequent NRF2 activation. With the increase of H2O2 levels under certain stresses and/or pathogenic conditions, H2O2 could reach levels that could activate the KEAP1-NRF2-ARE system in a concentration-dependent manner. However, following further increase of H2O2, the steady-state concentration of NRF2 will lose its ultrasensitivity, and then enter the plateau stage, at which further increased H2O2 cannot effectively increase nuclear NRF2 levels. Thus, it is evident that the distinct paradoxical roles of ROS in inflammatory response are concentration dependent.
RCD, including pyroptosis, ferroptosis, and apoptosis, involves tightly structured signaling cascades and molecularly defined effector mechanisms, in which ROS play an important part [69,70]. A growing number of novel forms of RCD have been identified and known as crucial factors in various pathophysiological settings. Among the key molecules regulating RCD, the nucleotide-binding domain, leucine-rich-repeat-containing family, pyrin domain-containing 3 (NLRP3) inflammasome is noticeable, because it is widely involved in a variety of pathogenic conditions [51,71]. In cells exposed to PAMPs and/or DAMPs, the NLRP3 inflammasome detects microbial products and stressors and subsequently activates caspase-1, 4, 5, and 11, leading to the overproduction of IL-1β, IL-18, and other DAMPs [72]. It has been well documented that the activation of NLRP3 is critical in pyroptosis and is deeply influenced by ROS [73,74,75]. In addition, ROS also serve as key activating factors in ferroptosis, a distinct RCD caused by the accumulation of lipid peroxides through iron-mediated lipid peroxidation [69,76], and apoptosis, which involves the genetically determined elimination of cells initiated by caspase-8 and 9 and executed by caspase-3, 6, and 7 [77]. Thus, overproduction of ROS is critically important in a variety of RCD, which may aggravate the release of DAMPs, leading to a pro-inflammatory response.
ROS can function as signaling molecules to initiate the primary inflammatory responses and the secondary damage in cytokine storms [78,79]. Increased levels of ROS initiate and maintain the abnormal activation of NF-κB and MAPK and the excess of pro-inflammatory cytokines, such as IL-6 [80,81,82]. IL-6, as a key DAMP produced by PAMPs- and/or DAMPs-activated macrophages, might bind with two different receptors, bringing about diametrically opposite effects [83]. On the one hand, IL-6 mostly drives the acute-phase response by binding to the soluble receptor and subsequently activating a variety of immune cells including T-cells. In turn, activated T-cells and natural killer (NK) cells produce IFN-γ and augment macrophage activation, constituting a positive feedback loop among the immune cells to sustain the inflammation environment and promote anti-pathogen immunity [84]. On the other hand, IL-6 may also trigger an anti-inflammatory response via binding to the membrane-bound IL-6 receptor (mIL-6R) [45,85]. In line with the critical roles of IL-6 in inflammation, TNF-α may stimulate mitochondrial ROS production and trigger inflammatory cell death, tissue damage, and mortality via binding to the membrane-bound or soluble receptors [86], and thus accelerate the ROS–cytokines vicious cycle.
The molecular details underlying the stimulatory effects of ROS on cytokine production are complicated. The ROS produced from NOX on the membrane interact directly with the canonical pathways by affecting the inhibitor of NF-κB (IκB) and IκB kinase (IKK) β or the activation of NF-κB-inducing kinase, the upstream kinase in the alternative pathway [87,88]. In addition, ROS can activate MAPK kinase and initiate the MAPK cascade [89,90]. Activation of the MAPKs leads to phosphorylation and activation of p38 present in the cytoplasm or nucleus, which initiates the inflammatory response. MAPKs are a family of protein kinases that respond to diverse stimuli by inducing pro-inflammatory cytokines (e.g., IL-1β, TNF-α, and IL-6) [91]. MAPKs also primarily respond to PAMPs, some of which, such as SARS-CoVs, could lead to an overabundant expression of the inflammatory mediators. In contrast, it has been established that the JAK-STAT signaling pathway functions as a key mediator in IFN-β and TNF-α-induced ROS production [92,93].
In summary, the primary inflammatory responses and systemic inflammatory effects in cytokine storms are closely associated with overproduced ROS, which may function as DAMPs to stimulate the production, maturation, and secretion of a variety of chemokines and pro-inflammatory cytokines. In turn, the chemokines and cytokines may activate and recruit more immune cells to generate more ROS, forming an amplification loop. Without enough antioxidants and/or anti-inflammatory factors to break the ROS–cytokines vicious cycle, severe inflammation and cytokine storms would happen (Figure 2).

4. NRF2 in Cytokine Storms

4.1. NRF2 Basics

As the master transcriptional factor in the cellular adaptive antioxidant response, NRF2 regulates the genes encoding many antioxidant and phase II detoxifying enzymes and cell metabolism-related factors, including γ-glutamate-cysteine ligase catalytic and modifier subunit (GCLC and GCLM), glutathione S-transferase (GST), heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidoreductase 1 (NQO1), and peroxisome proliferators-activated receptors-γ (PPAR-γ) [7,11,94]. Thus, NRF2-dependent transcription protects against oxidative damage and various redox-sensitive RCD by reducing ROS. In Nrf2-knockout mice, the damage to tissues and organs caused by oxidative stress is more significant than that in wild-type mice [95,96].

4.2. Key Cellular Events Regulated by NRF2 during Cytokine Storms

It has been well documented that distinct cellular events, including macrophage polarization, pyroptosis, endothelial cell damage, and neutrophil infiltration, are crucial in the development of cytokine storms, in which NRF2-mediated adaptive antioxidant response can alleviate their progress, to prevent deteriorated inflammation and thus cytokine storms (Figure 3).

4.2.1. Macrophage Polarization

Macrophages are complex immune cells that mainly have three phenotypes, M0, M1, and M2. Macrophage polarization, in which macrophages are converted from M0 to M1 or M2, or converted between M1 and M2, occurs in the initial stage of cytokine storms [97]. M1 macrophages are characterized by the production of high levels of pro-inflammatory cytokines, ROS, and reactive nitrogen species (RNS), as well as the induction of Type 1 helper T (Th1) responses [98]. In pathogen-induced cytokine storms, activation or polarization of innate immune cells mainly occur at the primarily infected loci, where M1 macrophages promote inflammation toward the subsequent stages of cytokine storms including multi-organ failure. In contrast, M2 macrophages are associated with anti-inflammatory responses, cell proliferation, and tissue repair [97,99].
Accumulating evidence indicates that NRF2 may function as a negative regulator of M1 polarization and a positive regulator of M2 polarization [100]. PAMPs and DAMPs, ROS in particular, are critical in the macrophage polarization toward the M1 phenotype by activating MAPK and NF-κB pathways [98]. In addition, PAMPs may also stimulate macrophages to overproduce ROS, which subsequently stimulate cytokine production. In contrast, decreased ROS levels are vital for M2 polarization [98,101].
HO-1 is a downstream target of NRF2 and is recognized as a cytoprotective enzyme via degrading heme into biliverdin, free iron, and carbon monoxide [98,102]. In sepsis models, NRF2-dependent induction of HO-1 is effective in decreasing M1 and enhancing M2 polarization by ROS scavenging and NF-κB suppression [103]. Thus, NRF2 can alleviate cytokine storms via ROS-dependent and independent mechanisms.

4.2.2. Pyroptosis

Pyroptosis is a form of RCD that occurs in parenchyma cells and inflammatory cells in response to pro-inflammatory stimuli [72]. Consequently, pyroptosis-derived DAMPs may result in the activation of inflammatory cascades in the primary loci and cause systemic inflammation. Given that ROS-mediated NLRP3 is the primary activator of caspase-1, NRF2 may alleviate pyroptosis via transcriptional regulation of various antioxidants [104,105,106,107]. Similarly, the activation of caspase-3 in target cells results in extensive pyroptosis in cytokine storms induced by CAR T-cell therapy [108]. Interestingly, NRF2 can also suppress pyroptosis through caspase-3 inhibition in unilateral ureteral obstruction [109]. Furthermore, pyroptosis suppression through the NRF2 cascade has been proved to prevent secondary organ injuries induced by cytokine storms [110].

4.2.3. Endothelial Dysfunction

Endothelial cells form the inner lining of the vessels and participate in the control of vascular permeability, the maintenance of blood fluidity, angiogenesis, and the trafficking of immune cells [111]. At the primary loci stage of cytokine storms, PAMPs and DAMPs released into the circulation can act on the endothelium to initiate RCD and/or alter their endocrine function to facilitate cytokine storms [2]. In turn, endothelial dysfunction contributes to systemic inflammation [112]. In virus and liposaccharide (LPS)-induced cytokine storms, NRF2 can prevent endothelial dysfunction by upregulating antioxidant enzymes, such as HO-1 and NQO-1, alleviating the death of endothelial cells [112,113]. By suppressing ROS, NRF2 can suppress a variety of RCD of endothelial cells, including ferroptosis, apoptosis, and pyroptosis [112,114,115]. PAMPs and subsequent DAMPs induce the release of NO, which mediates the relaxation of the blood vessels and the aggressive inflammatory symptoms by dilating the blood vessels [22]. Interestingly, NRF2 can effectively reduce NO production by upregulating antioxidant enzymes, including HO-1 [116].
Endothelial cells release chemokines to attract immune cells, and traffic them through the vascular wall [22,111], which further promotes endothelial permeability and induces multi-organ injuries [117]. NRF2 can suppress the recruitment of immune cells by inhibiting production of chemokines in a ROS-dependent fashion [118]. Under the action of PAMPs and/or DAMPs, endothelial cells can also produce cell adhesion molecules, including integrins, selectins, the immunoglobin superfamily, and cadherins, to recruit immune cells. NRF2 may downregulate these cell adhesion molecules by suppressing NF-κB in an antioxidant-dependent manner [113,119,120].
Endothelial cells are important in maintaining blood fluidity in normal conditions. Endothelial dysfunction is connected with coagulation disorders, including thromboembolism and disseminated intravascular coagulation (DIC) [121]. Thromboembolism and DIC are important pathological processes in cytokine storms. Coagulation disorders can contribute to vascular occlusion, hemorrhage, and tissue necrosis [2,121,122]. Endothelial dysfunction may cause the antithrombotic surface to be destroyed and coagulator leakage, which will finally cause a coagulation disorder [123]. These processes are regulated by ROS, and NRF2 can suppress ROS to alleviate these coagulation disorders [112].

4.2.4. Neutrophil Infiltration

In cytokine storms, neutrophils are the most important immune cells recruited by chemokines and cell adhesion molecules, leading to multi-organ injuries and increased endothelial permeability. Neutrophils also form NET, a three-dimensional web-like structure composed of DNA, histones, and granular proteins, to elicit an innate immune response by entrapping microorganisms [117,124]. In addition, NETs may amplify cytokine production in the primary response and exacerbate secondary injuries during cytokine storms [117]. Accumulating evidence indicates that NRF2 plays vital roles in regulating neutrophil recruitment in an antioxidants-dependent fashion [124,125,126]. By suppressing ROS, NRF2-dependent transcription can promote rolling velocity and reduce the adhesion to inhibit neutrophil recruitment [119,126].
Several studies have shown that NRF2 activators can decrease neutrophil infiltration and cytokine levels, including IL-1β, TNF-α, and IL-6, leading to the failure of NETs [119,124]. In addition, NRF2-mediated transcription of antioxidant enzymes reduces ROS levels and suppresses neutrophil production of IL-6 and TNF-α in sepsis models [127,128]. Furthermore, NRF2 has also been known to upregulate antioxidant enzymes in healthy neutrophils and reduce ROS, and alleviate the production of IL-6 and TNF-α in neutrophils from septic patients [129].
In summary, the NRF2-mediated antioxidant response can alleviate inflammatory responses via regulating many crucial cellular events during cytokine storms, including macrophage polarization, pyroptosis, endothelial cell damage, and neutrophil infiltration (Figure 3). It is of worth noting that the molecular mechanisms underlying the regulatory roles of NRF2 in the key cellular events during cytokine storms are complicated and warrant further detailed investigations.

4.3. NRF2 Regulates the Transcription and Maturation of Cytokines

In cytokine storms, the overexpression of cytokines is the most noticeable feature because they are the primary molecules connecting all the stages. Interestingly, NRF2 may modulate cytokine production and release, not only at the primary loci but also the systemic levels, in which the transcription and maturation of cytokines are sensitive to ROS accumulation [110,130]. As illustrated in Figure 4, NRF2 activation, on the one hand, downregulates the mRNA levels of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-1β, by inhibiting NF-κB activation [100,131,132]. In line with this concept, in the absence of NRF2, the mRNA levels of pro-inflammatory cytokines are much higher than those in wild-type conditions [12]. On the other hand, NRF2 positively affects the transcription of anti-inflammatory cytokines, such as IL-10 [133,134,135]. The mechanisms for NRF2 downregulating cytokine levels at the transcriptional level are complex. First, NRF2 can regulate cytokine transcription by acting directly on the DNA, in which NRF2 binds to the promoters of cytokine genes, including IL-6 and IL-1β, reducing their transcription [10,13]. Second, NRF2 can block the recruitment of RNA polymerase II (RNA Pol II), thereby suppressing the transcription process [10]. In contrast, it has also been demonstrated that NRF2 plays a positive role in IL-6 transcription by binding directly to the promoter [136,137], suggesting that NRF2 is critical in the fine tuning of IL-6 expression under complicated inflammatory conditions.
Cytokines are directly regulated by multiple transcription factors and posttranslational mechanisms, including NF-κB, MAPKs, and STAT. The activity of NF-κB can be affected by ROS, which are tightly controlled by antioxidant systems [88,89]. By reducing phosphorylation of IκBα, NRF2-dependent antioxidants can suppress NF-κB activation [138,139]. The phosphorylation of IκBα is catalyzed by IKKβ, which may also be activated by ROS. In addition, NRF2 suppresses NF-κB by upregulating downstream antioxidant enzymes, including superoxide dismutase (SOD), HO-1, and NQO1 [100,140,141]. In cytokine storms caused by sepsis and IAV infections, NRF2 activation results in reduced availability of ROS, NF-κB inhibition, and decreased cytokine production [142,143]. In addition, the mRNA and protein levels of NF-κB are downregulated by NRF2 activation in an antioxidant-dependent way [138,139].
Activation of MAPKs may also be suppressed by NRF2 activation [144]. ROS are capable of initiating MAPK cascades, in which each layer of kinase phosphorylates the lower layer and finally activates MAPK [145]. As a protein kinase, MAPK further activates transcription factors to increase the transcription of various cytokine genes [146]. In cytokine storms derived from sepsis or IAV infections, an NRF2-dependent antioxidant response reduces the phosphorylation of p38, ERK, and c-Jun N-terminal kinase (JNK), all of which are members of the MAPK family, and further suppresses cytokine production, including IL-1β, IL-6, TNF-α, and IL-8 [143,147,148]. In an inflammation model induced by ischemia–reperfusion, NRF2 showed anti-inflammatory effects by upregulating HO-1 and suppressing p38 in an antioxidant-dependent manner [149]. Thus, NRF2 may suppress MAPK-mediated production of cytokines by reducing ROS levels during cytokine storms.
Previous studies, including our own, have shown that NRF2 promoted PPAR-γ transcription by directly binding to its promoter in adipocytes and hepatocytes [150,151,152]. In acute pulmonary inflammation, NRF2 activates PPAR-γ but suppresses the activity of NF-κB [153]. In line with these findings, PPAR-γ has also been found to suppress sepsis-induced cardiac cell injury, a common secondary organ injury in cytokine storms, by suppressing NF-κB and MAPKs [154]. With regard to the molecular details on the crosstalk between PPAR-γ and NF-κB, PPAR-γ has been found to suppress NF-κB activity via binding directly to NF-κB and/or inhibiting its co-activators [155,156]. In addition, PPAR-γ may suppress the activation of NF-κB and MAPKs by suppressing ROS production [155,157]. Hence, it is reasonable to speculate that NRF2 may mitigate cytokine storms by activating PPAR-γ. Nevertheless, the mechanisms regarding the mitigating effects of NRF2-mediated PPAR-γ activation on cytokine storms still need further investigation.
In agreement with the consensus that NRF2 negatively affects NF-κB activation, KEAP1 has also been found to be involved in the regulation of NF-κB activity [158]. Our previous studies showed that KEAP1 silencing results in exaggerated NF-κB activation and augmented inflammatory responses in LPS-challenged macrophages [137]. With regard to the mechanisms underlying the regulation of KEAP1 on NF-kB, Lee et al. reported that KEAP1 directly binds to IKKβ, which is important in the activation of NF-κB, leading to ubiquitin–proteasome degradation of IKKβ and subsequent inhibition of NF-κB [159]. In addition, KEAP1 has also been found to enhance the degradation of IKKβ through the autophagy–lysosome pathway [160]. As NRF2 can positively regulate KEAP1 [161], it is reasonable to speculate that NRF2 suppresses NF-κB by upregulating KEAP1. However, the regulatory function of the NRF2–KEAP1 feedback loop on NF-κB in cytokine storms still lacks direct experimental evidence.
STAT3 is a transcriptional factor that can promote the transcription of cytokine genes, and therefore plays a crucial role in cytokine storms. NRF2 suppresses the STAT3 pathway in two different modes. Firstly, NRF2 can regulate STAT3 via an antioxidant-independent mechanism, in which NRF2 promotes the expression of SHP. SHP is a nuclear receptor that not only binds to phosphorylated STAT3 to suppress its activity [162], but also regulates TLR- and NLRP3-induced inflammasome activation [163]. Secondly, NRF2 suppresses STAT3 activation by inhibiting its phosphorylation, whereby NRF2 induces glutathione peroxidase 2 (GPx2) to suppress JAK1, a key kinase for STAT3 phosphorylation [164]. It is worth noting that there is scarce direct experimental evidence supporting the idea that NRF2 can suppress STAT3 to alleviate cytokine storms.
In addition to regulating the transcription of cytokines, NRF2 may also be involved in the maturation of cytokine proteins by inhibiting the activation of key proteins via ROS-dependent mechanisms. Inflammasomes participate in cytokine maturation by promoting the development of caspases, which can cleave IL-1β and IL-18 precursors into bioactive forms [77,165]. While NRF2 activation is associated with the decrease of NLRP3 and caspase-1 [110,166,167], the maturation of IL-1β is closely associated with the presence of caspase-1. NLRP3 inflammasome, which can be activated by ROS, plays a crucial role in processing the precursor of caspase-1 into its mature form [77,165]. NRF2 suppresses the LPS-induced activation of the NLRP3 inflammasome by ROS scavenging, further decreasing the production of caspase-1 and IL-1β [105]. In sepsis-induced cytokine storms, NRF2 initiates an anti-inflammatory process by suppressing inflammasome and caspase-1 and alleviating mitochondria dysfunction [167]. The same phenomenon is also observed in a sepsis-induced liver injury model [110]. Of note, the antioxidant enzymes HO-1, SOD, and NQO1 were elevated in both experiments, indicating that NRF2 suppresses protein modifications in an antioxidant-dependent way.
In summary, the expression and maturation of a variety of cytokines are tightly regulated by ROS-mediated or redox-sensitive mechanisms, in which NRF2 plays crucial roles in antioxidant-dependent and independent manners (Figure 4).

4.4. Strategies to Mitigate Cytokine Storms by Targeting NRF2

Emerging evidence suggests that NRF2 is a valuable target for the prevention and treatment of cytokine storms. Biopsies from COVID-19 patients showed that genes associated with the NRF2-dependent antioxidant response, such as NQO1, PPAR-γ and HO-1, were decreased [27,168,169], suggesting that NRF2 activation could be a therapeutic strategy for COVID-19. Many proven NRF2 activators, including dimethyl fumarate (DMF), sulforaphane (SFN), curcumin, and PB125®, have been applied in treating inflammation-related disorders [170]. In septic mice, DMF treatment can downregulate the levels of TNF-α and IL-6 in the brain [171]. An in vitro experiment proved that DMF could suppress the SARS-CoV-19-induced inflammatory response [168]. Pretreatment of septic mice with SFN decreased the levels of TNF-α and IL-6 in lung and brain tissues [119,172]. In addition, SFN treatment increased the survival rate of mice, and alleviates the multi-organ injury induced by sepsis, including lung, liver, and kidney [173,174]. Curcumin also showed a protective effect against IAV infection-induced lung injury by reducing the levels of TNF-α, IL-1β, and IL-6 in the tissues [147,175]. Recently, a clinical trial showed that nano-curcumin could combat cytokine storms in COVID-19 patients [176]. In addition to those well-documented NRF2 activators, some novel phytochemicals can activate NRF2 to alleviate inflammation. In bile duct ligation mice, pentoxifylline was reported to suppress inflammation by activating NRF2, and prevent liver cirrhosis [177]. Vincamine protects mice from LPS-induced lung injury by activating NRF2 [178]. Hydroalcoholic curry leaf extract has been found to suppress IL-1β, IL-6, and TNF-α, and activate NRF2 in acute pancreatitis mice [179]. Although more clinical trials are required, studies have clearly shown that NRF2 is a valuable target in mitigating cytokine storms.
As detailed above, NRF2 modulates cytokine levels mainly through antioxidant-dependent indirect ways, in which ROS serve as key mediators. Given that ROS and the ROS-sensitive redox microenvironment are crucial for the activation of multiple cytokine-producing signal cascades, including NF-κB and MAPK, ROS and oxidative stress have been recognized as key targets to control inflammation. However, ROS function as a double-edged sword in inflammation because they are needed by the immune system to remove pathogens. In addition, ROS may also function as signaling molecules, mediating many critical physiological processes and stress responses [180,181,182]. Thus, NRF2-mediated induction of antioxidants, on the one hand, may suppress ROS to alleviate inflammation-related tissue injuries. On the other hand, sustained NRF2 activation may lower the availability of physiological ROS, resulting in weakened antimicrobial abilities of macrophages and neutrophils and impaired redox signaling. In consideration of the paradoxical roles of ROS and NRF2-mediated adaptive responses in inflammatory responses, it is important to determine when, where, and how to modulate NRF2 to effectively mitigate inflammation and cytokine storms and avoid any potential off-target effects.
Cytokine storms result from complex interactions of a variety of cells and systems, in which the roles of activated NRF2 and its crosstalk with diverse signaling pathways are not entirely defined. For example, it is still unclear whether NRF2 activation is involved in immunoparalysis, a period or state of immunosuppression following an initial hyperinflammatory response in sepsis. Because immunoparalysis is related to a high risk of sepsis and organ failures, the application of NRF2 activators to downregulate the immune system should be performed under constant surveillance to prevent exacerbations of infections. When facing immunoparalysis in particular, the NRF2 activation strategy should be considered with more caution. At the early stage of the acute response phase, proper activation of NRF2 could be considered an effective anti-inflammatory strategy. In contrast, the effectiveness of NRF2 activators in later stages is in doubt, because NRF2 has also been shown to have various pro-inflammatory effects [183].
In summary, the spatiotemporal roles of NRF2 in cytokine storms still need more detailed research to support the development of new intervention strategies. In addition, novel NRF2 activators with high activity and specificity are required to facilitate their potential applications in treating cytokine storms. Clearly, early surveillance and corresponding measures are needed to improve the prognosis of patients when NRF2 modulators are used to prevent and/or treat cytokine storms.

5. Conclusions and Perspectives

NRF2 is critical in regulating the fundamental cellular, transcriptional, and maturation events of cytokine storms. Thus, it is reasonable to consider NRF2 as a potential therapeutic target for various inflammation-related disorders. The challenges for controlling cytokine storms lie in dissecting the exact physiological and pathophysiological regulations of the inflammatory response. Expanding knowledge on NRF2-ARE signaling during inflammation would advance our understanding of cytokine storms and reveal potential applications in drug discovery. Given that ROS and NRF2 play a dual Yin–Yang role in a wide range of physiological and pathophysiological processes, NRF2, as the master regulator in charge of transcriptional control of many key antioxidant enzymes, may play paradoxical roles in redox signaling and cellular homeostasis. Depending on the nature of pathophysiological conditions, NRF2 could be the good, the bad, and the ugly. Suggested areas for future study include the molecular mechanisms of cytokine production and release and the spatiotemporal roles of NRF2 in the context of cytokine storms, including therapeutic settings. We anticipate that significant advances will be made in the near future to expand our understanding of NRF2 signaling pathways and to develop new drugs for cytokine storm-related diseases.

Author Contributions

Writing—original draft preparation, Z.L., P.D. and H.W.; writing—review and editing, J.P., H.W., S.L., Q.Z., Y.B. and Y.X.; supervision, J.P. and H.W.; project administration, J.P. and H.W.; funding acquisition, J.P., H.W. and Y.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China 81830099 (J.P.), 82020108027 (J.P.), 82073513 (H.W.) and 82022063 (Y.X.); National Key R&D Program of China of the Ministry of Science and Technology of the People’s Republic of China #2018YFC1311600 (J.P.); Natural Science Foundation of Liaoning Province 2022-MS-215 (H.W.); Shenyang Science and Technology Bureau of Support Program for Young Innovation Scholar RC210296 (H.W.); Innovation Team Support from China Medical University (CXTD2022004); High-level Talents Support Foundation from China Medical University 2400022052 (H.W.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the article.

Acknowledgments

Figure 1, Figure 2, and Figure 4 were created with BioRender.com.

Conflicts of Interest

The authors declare that they have no conflict of interest.

Abbreviations

ARE: antioxidant response element; bZIP: basic region-leucine zipper; CAR: chi-meric antigen receptor; COVID-19: coronavirus disease-19; Cul3: Cullin 3; DAMPs: damage-associated molecular patterns; DIC: disseminated intravascular coagulation; DMF: dimethyl fumarate; ERK: extracellular regulated protein kinases; GCLC: glutamate–cysteine ligase catalytic subunit; GCLM: glutamate–cysteine ligase modifier subunit; GPx2: glutathione peroxidase 2; GST: glutathione S-transferase; HO-1: Heme oxygenase-1; IAV: influenza A virus; IFN: interferon; IL: interleukin; IκB: inhibitor of NF-κB; IKK: IκB kinase; JAK-STAT: Janus kinase/signal transducer and activators of transcription; JNK: c-Jun N-terminal kinase; KEAP1: Kelch-like ECH-associated protein 1; LPS: lipopolysaccharide; MAPK: mitogen-activated protein kinase; mIL-6R: membrane-bound IL-6 receptor; NETs: neutrophil extracellular traps; NF-κB: nuclear factor kappa-light chain-enhancer of activated B; NK: nature kill; NLRP3: nucleotide-binding domain, leucine-rich-repeat-containing family, pyrin domain-containing 3; NOD2: nucleotide-binding oligomerization domain-containing protein 2; NOX: NADPH oxidase; NQO1: NAD(P)H: quinone oxidoreductase 1; NRF2: nuclear factor erythroid 2-related factor 2; PAMPs: pathogen-associated molecular patterns; PPAR-γ: peroxisome proliferators-activated receptor-γ; PRRs: pattern recognition receptors; RCD: regulated cell death; RNS: reactive nitrogen species; ROS: reactive oxygen species; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SFN: sulforaphane; SHP: small heterodimer protein; SOD: superoxide dismutase; TLRs: Toll-like receptors; TNF-α: tumor necrosis factor α; Th1: Type 1 helper T.

References

  1. Ferrara, J.L.; Abhyankar, S.; Gilliland, D.G. Cytokine storm of graft-versus-host disease: A critical effector role for interleukin-1. Transplant. Proc. 1993, 25, 1216–1217. [Google Scholar]
  2. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273. [Google Scholar] [CrossRef] [PubMed]
  3. Kim, J.S.; Lee, J.Y.; Yang, J.W.; Lee, K.H.; Effenberger, M.; Szpirt, W.; Kronbichler, A.; Shin, J.I. Immunopathogenesis and treatment of cytokine storm in COVID-19. Theranostics 2021, 11, 316–329. [Google Scholar] [CrossRef] [PubMed]
  4. Tang, X.D.; Ji, T.T.; Dong, J.R.; Feng, H.; Chen, F.Q.; Chen, X.; Zhao, H.Y.; Chen, D.K.; Ma, W.T. Pathogenesis and Treatment of Cytokine Storm Induced by Infectious Diseases. Int. J. Mol. Sci. 2021, 22, 13009. [Google Scholar] [CrossRef] [PubMed]
  5. Taguchi, K.; Yamamoto, M. The KEAP1-NRF2 System in Cancer. Front. Oncol. 2017, 7, 85. [Google Scholar] [CrossRef] [Green Version]
  6. Li, R.; Jia, Z.; Zhu, H. Regulation of Nrf2 Signaling. React. Oxyg. Species 2019, 8, 312–322. [Google Scholar] [CrossRef]
  7. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [Green Version]
  8. Liu, S.; Pi, J.; Zhang, Q. Signal amplification in the KEAP1-NRF2-ARE antioxidant response pathway. Redox Biol. 2022, 54, 102389. [Google Scholar] [CrossRef]
  9. Zhang, M.; An, C.; Gao, Y.; Leak, R.K.; Chen, J.; Zhang, F. Emerging roles of Nrf2 and phase II antioxidant enzymes in neuroprotection. Prog. Neurobiol. 2013, 100, 30–47. [Google Scholar] [CrossRef] [Green Version]
  10. Kobayashi, E.H.; Suzuki, T.; Funayama, R.; Nagashima, T.; Hayashi, M.; Sekine, H.; Tanaka, N.; Moriguchi, T.; Motohashi, H.; Nakayama, K.; et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat. Commun. 2016, 7, 11624. [Google Scholar] [CrossRef] [Green Version]
  11. Bellezza, I.; Giambanco, I.; Minelli, A.; Donato, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef] [PubMed]
  12. Pan, H.; Wang, H.; Zhu, L.; Mao, L.; Qiao, L.; Su, X. Depletion of Nrf2 enhances inflammation induced by oxyhemoglobin in cultured mice astrocytes. Neurochem. Res. 2011, 36, 2434–2441. [Google Scholar] [CrossRef] [PubMed]
  13. Early, J.O.; Menon, D.; Wyse, C.A.; Cervantes-Silva, M.P.; Zaslona, Z.; Carroll, R.G.; Palsson-McDermott, E.M.; Angiari, S.; Ryan, D.G.; Corcoran, S.E.; et al. Circadian clock protein BMAL1 regulates IL-1beta in macrophages via NRF2. Proc. Natl. Acad. Sci. USA 2018, 115, E8460–E8468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jhang, J.J.; Cheng, Y.T.; Ho, C.Y.; Yen, G.C. Monosodium urate crystals trigger Nrf2- and heme oxygenase-1-dependent inflammation in THP-1 cells. Cell. Mol. Immunol. 2015, 12, 424–434. [Google Scholar] [CrossRef] [Green Version]
  15. Khan, N.M.; Haseeb, A.; Ansari, M.Y.; Devarapalli, P.; Haynie, S.; Haqqi, T.M. Wogonin, a plant derived small molecule, exerts potent anti-inflammatory and chondroprotective effects through the activation of ROS/ERK/Nrf2 signaling pathways in human Osteoarthritis chondrocytes. Free Radic. Biol. Med. 2017, 106, 288–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Tisoncik, J.R.; Korth, M.J.; Simmons, C.P.; Farrar, J.; Martin, T.R.; Katze, M.G. Into the eye of the cytokine storm. Microbiol. Mol. Biol. Rev. 2012, 76, 16–32. [Google Scholar] [CrossRef] [Green Version]
  17. Link, H. The cytokine storm in multiple sclerosis. Mult. Scler. 1998, 4, 12–15. [Google Scholar] [CrossRef]
  18. Oldstone, M.B.; Rosen, H. Cytokine storm plays a direct role in the morbidity and mortality from influenza virus infection and is chemically treatable with a single sphingosine-1-phosphate agonist molecule. Curr. Top. Microbiol. Immunol. 2014, 378, 129–147. [Google Scholar] [CrossRef]
  19. Hantoushzadeh, S.; Norooznezhad, A.H. Possible Cause of Inflammatory Storm and Septic Shock in Patients Diagnosed with (COVID-19). Arch. Med. Res. 2020, 51, 347–348. [Google Scholar] [CrossRef]
  20. Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. J. Am. Soc. Gene Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef]
  21. Karki, R.; Sharma, B.R.; Tuladhar, S.; Williams, E.P.; Zalduondo, L.; Samir, P.; Zheng, M.; Sundaram, B.; Banoth, B.; Malireddi, R.K.S.; et al. Synergism of TNF-alpha and IFN-gamma Triggers Inflammatory Cell Death, Tissue Damage, and Mortality in SARS-CoV-2 Infection and Cytokine Shock Syndromes. Cell 2021, 184, 149–168.e17. [Google Scholar] [CrossRef]
  22. Chousterman, B.G.; Swirski, F.K.; Weber, G.F. Cytokine storm and sepsis disease pathogenesis. Semin. Immunopathol. 2017, 39, 517–528. [Google Scholar] [CrossRef] [PubMed]
  23. Godfred-Cato, S.; Bryant, B.; Leung, J.; Oster, M.E.; Conklin, L.; Abrams, J.; Roguski, K.; Wallace, B.; Prezzato, E.; Koumans, E.H.; et al. COVID-19-Associated Multisystem Inflammatory Syndrome in Children—United States, March-July 2020. MMWR Morb. Mortal. Wkl. Rep. 2020, 69, 1074–1080. [Google Scholar] [CrossRef] [PubMed]
  24. Morris, S.B.; Schwartz, N.G.; Patel, P.; Abbo, L.; Beauchamps, L.; Balan, S.; Lee, E.H.; Paneth-Pollak, R.; Geevarughese, A.; Lash, M.K.; et al. Case Series of Multisystem Inflammatory Syndrome in Adults Associated with SARS-CoV-2 Infection—United Kingdom and United States, March-August 2020. MMWR Morb. Mortal. Wkl. Rep. 2020, 69, 1450–1456. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed] [Green Version]
  26. Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M.; Ellingson, M.K.; Mao, T.; Oh, J.E.; Israelow, B.; et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469. [Google Scholar] [CrossRef] [PubMed]
  27. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
  28. Wu, H.P.; Chen, C.K.; Chung, K.; Tseng, J.C.; Hua, C.C.; Liu, Y.C.; Chuang, D.Y.; Yang, C.H. Serial cytokine levels in patients with severe sepsis. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2009, 58, 385–393. [Google Scholar] [CrossRef]
  29. Giavridis, T.; van der Stegen, S.J.C.; Eyquem, J.; Hamieh, M.; Piersigilli, A.; Sadelain, M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat. Med. 2018, 24, 731–738. [Google Scholar] [CrossRef]
  30. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet 2015, 385, 517–528. [Google Scholar] [CrossRef] [PubMed]
  31. Stella, A.; Lamkanfi, M.; Portincasa, P. Familial Mediterranean Fever and COVID-19: Friends or Foes? Front. Immunol. 2020, 11, 574593. [Google Scholar] [CrossRef]
  32. Savic, S.; Dickie, L.J.; Wittmann, M.; McDermott, M.F. Autoinflammatory syndromes and cellular responses to stress: Pathophysiology, diagnosis and new treatment perspectives. Best Pract. Res. Clin. Rheumatol. 2012, 26, 505–533. [Google Scholar] [CrossRef] [PubMed]
  33. Xu, X.J.; Tang, Y.M.; Song, H.; Yang, S.L.; Xu, W.Q.; Zhao, N.; Shi, S.W.; Shen, H.P.; Mao, J.Q.; Zhang, L.Y.; et al. Diagnostic accuracy of a specific cytokine pattern in hemophagocytic lymphohistiocytosis in children. J. Pediatr. 2012, 160, 984–990.e1. [Google Scholar] [CrossRef] [PubMed]
  34. Wrobleski, D.M.; Barth, M.M.; Oyen, L.J. Necrotizing pancreatitis: Pathophysiology, diagnosis, and acute care management. AACN Clin. Issues 1999, 10, 464–477. [Google Scholar] [CrossRef]
  35. Hegyi, P.; Szakacs, Z.; Sahin-Toth, M. Lipotoxicity and Cytokine Storm in Severe Acute Pancreatitis and COVID-19. Gastroenterology 2020, 159, 824–827. [Google Scholar] [CrossRef]
  36. Savarin, C.; Bergmann, C.C. Fine Tuning the Cytokine Storm by IFN and IL-10 Following Neurotropic Coronavirus Encephalomyelitis. Front. Immunol. 2018, 9, 3022. [Google Scholar] [CrossRef] [Green Version]
  37. de Candia, P.; Prattichizzo, F.; Garavelli, S.; Matarese, G. T Cells: Warriors of SARS-CoV-2 Infection. Trends Immunol. 2021, 42, 18–30. [Google Scholar] [CrossRef]
  38. Cagliero, J.; Villanueva, S.; Matsui, M. Leptospirosis Pathophysiology: Into the Storm of Cytokines. Front. Cell. Infect. Microbiol. 2018, 8, 204. [Google Scholar] [CrossRef] [Green Version]
  39. Lee, S.; Channappanavar, R.; Kanneganti, T.D. Coronaviruses: Innate Immunity, Inflammasome Activation, Inflammatory Cell Death, and Cytokines. Trends Immunol. 2020, 41, 1083–1099. [Google Scholar] [CrossRef]
  40. Spaulding, A.R.; Salgado-Pabon, W.; Kohler, P.L.; Horswill, A.R.; Leung, D.Y.; Schlievert, P.M. Staphylococcal and streptococcal superantigen exotoxins. Clin. Microbiol. Rev. 2013, 26, 422–447. [Google Scholar] [CrossRef] [Green Version]
  41. Karki, R.; Kanneganti, T.D. The ‘cytokine storm’: Molecular mechanisms and therapeutic prospects. Trends Immunol. 2021, 42, 681–705. [Google Scholar] [CrossRef]
  42. Yang, Y.; Tang, H. Aberrant coagulation causes a hyper-inflammatory response in severe influenza pneumonia. Cell. Mol. Immunol. 2016, 13, 432–442. [Google Scholar] [CrossRef] [Green Version]
  43. Krakauer, T. Staphylococcal Superantigens: Pyrogenic Toxins Induce Toxic Shock. Toxins 2019, 11, 178. [Google Scholar] [CrossRef] [Green Version]
  44. Janeway, C.A., Jr.; Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef] [Green Version]
  45. Fara, A.; Mitrev, Z.; Rosalia, R.A.; Assas, B.M. Cytokine storm and COVID-19: A chronicle of pro-inflammatory cytokines. Open Biol. 2020, 10, 200160. [Google Scholar] [CrossRef]
  46. Wang, H.; Ma, S. The cytokine storm and factors determining the sequence and severity of organ dysfunction in multiple organ dysfunction syndrome. Am. J. Emerg. Med. 2008, 26, 711–715. [Google Scholar] [CrossRef]
  47. Kurahashi, K.; Kajikawa, O.; Sawa, T.; Ohara, M.; Gropper, M.A.; Frank, D.W.; Martin, T.R.; Wiener-Kronish, J.P. Pathogenesis of septic shock in Pseudomonas aeruginosa pneumonia. J. Clin. Investig. 1999, 104, 743–750. [Google Scholar] [CrossRef] [Green Version]
  48. Pelaia, C.; Tinello, C.; Vatrella, A.; De Sarro, G.; Pelaia, G. Lung under attack by COVID-19-induced cytokine storm: Pathogenic mechanisms and therapeutic implications. Ther. Adv. Respir. Dis. 2020, 14, 1753466620933508. [Google Scholar] [CrossRef]
  49. Abbasifard, M.; Khorramdelazad, H. The bio-mission of interleukin-6 in the pathogenesis of COVID-19: A brief look at potential therapeutic tactics. Life Sci. 2020, 257, 118097. [Google Scholar] [CrossRef]
  50. 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. Intensiv. Care Med. 2020, 46, 846–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Freeman, T.L.; Swartz, T.H. Targeting the NLRP3 Inflammasome in Severe COVID-19. Front. Immunol. 2020, 11, 1518. [Google Scholar] [CrossRef] [PubMed]
  52. Hwang, J.; Jin, J.; Jeon, S.; Moon, S.H.; Park, M.Y.; Yum, D.Y.; Kim, J.H.; Kang, J.E.; Park, M.H.; Kim, E.J.; et al. SOD1 suppresses pro-inflammatory immune responses by protecting against oxidative stress in colitis. Redox Biol. 2020, 37, 101760. [Google Scholar] [CrossRef]
  53. Lee, W.; Ahn, J.H.; Park, H.H.; Kim, H.N.; Kim, H.; Yoo, Y.; Shin, H.; Hong, K.S.; Jang, J.G.; Park, C.G.; et al. COVID-19-activated SREBP2 disturbs cholesterol biosynthesis and leads to cytokine storm. Signal Transduct. Target. Ther. 2020, 5, 186. [Google Scholar] [CrossRef]
  54. Hur, S. Double-Stranded RNA Sensors and Modulators in Innate Immunity. Annu. Rev. Immunol. 2019, 37, 349–375. [Google Scholar] [CrossRef]
  55. Lazear, H.M.; Schoggins, J.W.; Diamond, M.S. Shared and Distinct Functions of Type I and Type III Interferons. Immunity 2019, 50, 907–923. [Google Scholar] [CrossRef] [PubMed]
  56. Banerjee, S.; Biehl, A.; Gadina, M.; Hasni, S.; Schwartz, D.M. JAK-STAT Signaling as a Target for Inflammatory and Autoimmune Diseases: Current and Future Prospects. Drugs 2017, 77, 521–546. [Google Scholar] [CrossRef]
  57. Kang, S.; Tanaka, T.; Narazaki, M.; Kishimoto, T. Targeting Interleukin-6 Signaling in Clinic. Immunity 2019, 50, 1007–1023. [Google Scholar] [CrossRef]
  58. Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes 2003, 52, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Droge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef] [Green Version]
  60. Hansen, J.M.; Go, Y.M.; Jones, D.P. Nuclear and mitochondrial compartmentation of oxidative stress and redox signaling. Annu. Rev. Pharmacol. Toxicol. 2006, 46, 215–234. [Google Scholar] [CrossRef]
  61. Liu, P.; Wang, X.; Sun, Y.; Zhao, H.; Cheng, F.; Wang, J.; Yang, F.; Hu, J.; Zhang, H.; Wang, C.C.; et al. SARS-CoV-2 ORF8 reshapes the ER through forming mixed disulfides with ER oxidoreductases. Redox Biol. 2022, 54, 102388. [Google Scholar] [CrossRef]
  62. Taylor, J.P.; Tse, H.M. The role of NADPH oxidases in infectious and inflammatory diseases. Redox Biol. 2021, 48, 102159. [Google Scholar] [CrossRef]
  63. Herb, M.; Schramm, M. Functions of ROS in Macrophages and Antimicrobial Immunity. Antioxidants 2021, 10, 313. [Google Scholar] [CrossRef]
  64. Cobley, J.N. Mechanisms of Mitochondrial ROS Production in Assisted Reproduction: The Known, the Unknown, and the Intriguing. Antioxidants 2020, 9, 933. [Google Scholar] [CrossRef]
  65. Dispenzieri, A.; Fajgenbaum, D.C. Overview of Castleman disease. Blood 2020, 135, 1353–1364. [Google Scholar] [CrossRef]
  66. Kumar, V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int. Immunopharmacol. 2020, 89, 107087. [Google Scholar] [CrossRef]
  67. Shirey, K.A.; Blanco, J.C.G.; Vogel, S.N. Targeting TLR4 Signaling to Blunt Viral-Mediated Acute Lung Injury. Front. Immunol. 2021, 12, 705080. [Google Scholar] [CrossRef]
  68. Liu, S.; Pi, J.; Zhang, Q. Mathematical modeling reveals quantitative properties of KEAP1-NRF2 signaling. Redox Biol. 2021, 47, 102139. [Google Scholar] [CrossRef]
  69. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef] [Green Version]
  70. Benhar, M. Oxidants, Antioxidants and Thiol Redox Switches in the Control of Regulated Cell Death Pathways. Antioxidants 2020, 9, 309. [Google Scholar] [CrossRef] [Green Version]
  71. Satoh, T.; Trudler, D.; Oh, C.K.; Lipton, S.A. Potential Therapeutic Use of the Rosemary Diterpene Carnosic Acid for Alzheimer’s Disease, Parkinson’s Disease, and Long-COVID through NRF2 Activation to Counteract the NLRP3 Inflammasome. Antioxidants 2022, 11, 124. [Google Scholar] [CrossRef]
  72. Vande Walle, L.; Lamkanfi, M. Pyroptosis. Curr. Biol. CB 2016, 26, R568–R572. [Google Scholar] [CrossRef] [Green Version]
  73. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  74. Pang, Y.; Wu, D.; Ma, Y.; Cao, Y.; Liu, Q.; Tang, M.; Pu, Y.; Zhang, T. Reactive oxygen species trigger NF-kappaB-mediated NLRP3 inflammasome activation involvement in low-dose CdTe QDs exposure-induced hepatotoxicity. Redox Biol. 2021, 47, 102157. [Google Scholar] [CrossRef]
  75. Dan Dunn, J.; 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]
  76. Fakhouri, E.W.; Peterson, S.J.; Kothari, J.; Alex, R.; Shapiro, J.I.; Abraham, N.G. Genetic Polymorphisms Complicate COVID-19 Therapy: Pivotal Role of HO-1 in Cytokine Storm. Antioxidants 2020, 9, 636. [Google Scholar] [CrossRef]
  77. Van Opdenbosch, N.; Lamkanfi, M. Caspases in Cell Death, Inflammation, and Disease. Immunity 2019, 50, 1352–1364. [Google Scholar] [CrossRef]
  78. Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [Green Version]
  79. Codo, A.C.; Davanzo, G.G.; Monteiro, L.B.; de Souza, G.F.; Muraro, S.P.; Virgilio-da-Silva, J.V.; Prodonoff, J.S.; Carregari, V.C.; de Biagi Junior, C.A.O.; Crunfli, F.; et al. Elevated Glucose Levels Favor SARS-CoV-2 Infection and Monocyte Response through a HIF-1alpha/Glycolysis-Dependent Axis. Cell Metab. 2020, 32, 437–446.e5. [Google Scholar] [CrossRef]
  80. Bulua, A.C.; Simon, A.; Maddipati, R.; Pelletier, M.; Park, H.; Kim, K.Y.; Sack, M.N.; Kastner, D.L.; Siegel, R.M. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J. Exp. Med. 2011, 208, 519–533. [Google Scholar] [CrossRef]
  81. Sengupta, P.; Dutta, S.; Roychoudhury, S.; D’Souza, U.J.A.; Govindasamy, K.; Kolesarova, A. COVID-19, Oxidative Stress and Male Reproduction: Possible Role of Antioxidants. Antioxidants 2022, 11, 548. [Google Scholar] [CrossRef] [PubMed]
  82. Ramos-Tovar, E.; Muriel, P. Molecular Mechanisms That Link Oxidative Stress, Inflammation, and Fibrosis in the Liver. Antioxidants 2020, 9, 1279. [Google Scholar] [CrossRef]
  83. Del Valle, D.M.; Kim-Schulze, S.; Huang, H.H.; Beckmann, N.D.; Nirenberg, S.; Wang, B.; Lavin, Y.; Swartz, T.H.; Madduri, D.; Stock, A.; et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat. Med. 2020, 26, 1636–1643. [Google Scholar] [CrossRef]
  84. Vadhan-Raj, S.; Nathan, C.F.; Sherwin, S.A.; Oettgen, H.F.; Krown, S.E. Phase I trial of recombinant interferon gamma by 1-hour i.v. infusion. Cancer Treat. Rep. 1986, 70, 609–614. [Google Scholar]
  85. Paniri, A.; Akhavan-Niaki, H. Emerging role of IL-6 and NLRP3 inflammasome as potential therapeutic targets to combat COVID-19: Role of lncRNAs in cytokine storm modulation. Life Sci. 2020, 257, 118114. [Google Scholar] [CrossRef]
  86. Blaser, H.; Dostert, C.; Mak, T.W.; Brenner, D. TNF and ROS Crosstalk in Inflammation. Trends Cell Biol. 2016, 26, 249–261. [Google Scholar] [CrossRef] [PubMed]
  87. Korbecki, J.; Baranowska-Bosiacka, I.; Gutowska, I.; Chlubek, D. The effect of reactive oxygen species on the synthesis of prostanoids from arachidonic acid. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 2013, 64, 409–421. [Google Scholar]
  88. Lawrence, T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 2009, 1, a001651. [Google Scholar] [CrossRef] [Green Version]
  89. Sun, J.; Wang, J.; Li, L.; Wu, Z.; Chen, X.; Yuan, J. ROS induced by spring viraemia of carp virus activate the inflammatory response via the MAPK/AP-1 and PI3K signaling pathways. Fish Shellfish Immunol. 2020, 101, 216–224. [Google Scholar] [CrossRef]
  90. Zhang, Y.; Yang, S.; Qiu, Z.; Huang, L.; Huang, L.; Liang, Y.; Liu, X.; Wang, M.; Zhou, B. Pyrogallol enhances therapeutic effect of human umbilical cord mesenchymal stem cells against LPS-mediated inflammation and lung injury via activation of Nrf2/HO-1 signaling. Free Radic. Biol. Med. 2022, 191, 66–81. [Google Scholar] [CrossRef]
  91. Yu, J.; Sun, X.; Goie, J.Y.G.; Zhang, Y. Regulation of Host Immune Responses against Influenza A Virus Infection by Mitogen-Activated Protein Kinases (MAPKs). Microorganisms 2020, 8, 1067. [Google Scholar] [CrossRef] [PubMed]
  92. Abroun, S.; Saki, N.; Ahmadvand, M.; Asghari, F.; Salari, F.; Rahim, F. STATs: An Old Story, Yet Mesmerizing. Cell J. 2015, 17, 395–411. [Google Scholar] [CrossRef] [PubMed]
  93. Fink, K.; Martin, L.; Mukawera, E.; Chartier, S.; De Deken, X.; Brochiero, E.; Miot, F.; Grandvaux, N. IFNbeta/TNFalpha synergism induces a non-canonical STAT2/IRF9-dependent pathway triggering a novel DUOX2 NADPH oxidase-mediated airway antiviral response. Cell Res. 2013, 23, 673–690. [Google Scholar] [CrossRef]
  94. Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem. Biophys. Res. Commun. 1997, 236, 313–322. [Google Scholar] [CrossRef]
  95. Kovac, S.; Angelova, P.R.; Holmstrom, K.M.; Zhang, Y.; Dinkova-Kostova, A.T.; Abramov, A.Y. Nrf2 regulates ROS production by mitochondria and NADPH oxidase. Biochim. Biophys. Acta 2015, 1850, 794–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Crilly, M.J.; Tryon, L.D.; Erlich, A.T.; Hood, D.A. The role of Nrf2 in skeletal muscle contractile and mitochondrial function. J. Appl. Physiol. 2016, 121, 730–740. [Google Scholar] [CrossRef] [Green Version]
  97. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  98. Tan, H.Y.; Wang, N.; Li, S.; Hong, M.; Wang, X.; Feng, Y. The Reactive Oxygen Species in Macrophage Polarization: Reflecting Its Dual Role in Progression and Treatment of Human Diseases. Oxid. Med. Cell. Longev. 2016, 2016, 2795090. [Google Scholar] [CrossRef] [Green Version]
  99. Lallemand, T.; Rouahi, M.; Swiader, A.; Grazide, M.H.; Geoffre, N.; Alayrac, P.; Recazens, E.; Coste, A.; Salvayre, R.; Negre-Salvayre, A.; et al. nSMase2 (Type 2-Neutral Sphingomyelinase) Deficiency or Inhibition by GW4869 Reduces Inflammation and Atherosclerosis in Apoe(-/-) Mice. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1479–1492. [Google Scholar] [CrossRef] [Green Version]
  100. Qin, T.; Du, R.; Huang, F.; Yin, S.; Yang, J.; Qin, S.; Cao, W. Sinomenine activation of Nrf2 signaling prevents hyperactive inflammation and kidney injury in a mouse model of obstructive nephropathy. Free Radic. Biol. Med. 2016, 92, 90–99. [Google Scholar] [CrossRef]
  101. Griess, B.; Mir, S.; Datta, K.; Teoh-Fitzgerald, M. Scavenging reactive oxygen species selectively inhibits M2 macrophage polarization and their pro-tumorigenic function in part, via Stat3 suppression. Free Radic. Biol. Med. 2020, 147, 48–60. [Google Scholar] [CrossRef]
  102. Naito, Y.; Takagi, T.; Higashimura, Y. Heme oxygenase-1 and anti-inflammatory M2 macrophages. Arch. Biochem. Biophys. 2014, 564, 83–88. [Google Scholar] [CrossRef] [Green Version]
  103. Shen, K.; Jia, Y.; Wang, X.; Zhang, J.; Liu, K.; Wang, J.; Cai, W.; Li, J.; Li, S.; Zhao, M.; et al. Exosomes from adipose-derived stem cells alleviate the inflammation and oxidative stress via regulating Nrf2/HO-1 axis in macrophages. Free Radic. Biol. Med. 2021, 165, 54–66. [Google Scholar] [CrossRef] [PubMed]
  104. Fei, L.; Jingyuan, X.; Fangte, L.; Huijun, D.; Liu, Y.; Ren, J.; Jinyuan, L.; Linghui, P. Preconditioning with rHMGB1 ameliorates lung ischemia-reperfusion injury by inhibiting alveolar macrophage pyroptosis via the Keap1/Nrf2/HO-1 signaling pathway. J. Transl. Med. 2020, 18, 301. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, X.; Zhang, X.; Ding, Y.; Zhou, W.; Tao, L.; Lu, P.; Wang, Y.; Hu, R. Nuclear Factor E2-Related Factor-2 Negatively Regulates NLRP3 Inflammasome Activity by Inhibiting Reactive Oxygen Species-Induced NLRP3 Priming. Antioxid. Redox Signal. 2017, 26, 28–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Yu, J.; Chen, J.; Yang, H.; Chen, S.; Wang, Z. Overexpression of miR-200a-3p promoted inflammation in sepsis-induced brain injury through ROS-induced NLRP3. Int. J. Mol. Med. 2019, 44, 1811–1823. [Google Scholar] [CrossRef]
  107. Yang, W.; Wang, Y.; Zhang, P.; Sun, X.; Chen, X.; Yu, J.; Shi, L.; Yin, Y.; Tao, K.; Li, R. Immune-responsive gene 1 protects against liver injury caused by concanavalin A via the activation Nrf2/HO-1 pathway and inhibition of ROS activation pathways. Free Radic. Biol. Med. 2022, 182, 108–118. [Google Scholar] [CrossRef]
  108. Liu, Y.; Fang, Y.; Chen, X.; Wang, Z.; Liang, X.; Zhang, T.; Liu, M.; Zhou, N.; Lv, J.; Tang, K.; et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci. Immunol. 2020, 5, eaax7969. [Google Scholar] [CrossRef]
  109. Chung, S.D.; Lai, T.Y.; Chien, C.T.; Yu, H.J. Activating Nrf-2 signaling depresses unilateral ureteral obstruction-evoked mitochondrial stress-related autophagy, apoptosis and pyroptosis in kidney. PLoS ONE 2012, 7, e47299. [Google Scholar] [CrossRef]
  110. Hong, M.K.; Hu, L.L.; Zhang, Y.X.; Xu, Y.L.; Liu, X.Y.; He, P.K.; Jia, Y.H. 6-Gingerol ameliorates sepsis-induced liver injury through the Nrf2 pathway. Int. Immunopharmacol. 2020, 80, 106196. [Google Scholar] [CrossRef]
  111. Aird, W.C. Endothelial cell heterogeneity. Cold Spring Harb. Perspect. Med. 2012, 2, a006429. [Google Scholar] [CrossRef] [Green Version]
  112. Shao, Y.; Saredy, J.; Xu, K.; Sun, Y.; Saaoud, F.; Drummer, C.T.; Lu, Y.; Luo, J.J.; Lopez-Pastrana, J.; Choi, E.T.; et al. Endothelial Immunity Trained by Coronavirus Infections, DAMP Stimulations and Regulated by Anti-Oxidant NRF2 May Contribute to Inflammations, Myelopoiesis, COVID-19 Cytokine Storms and Thromboembolism. Front. Immunol. 2021, 12, 653110. [Google Scholar] [CrossRef]
  113. Chen, H.; Xie, K.; Han, H.; Li, Y.; Liu, L.; Yang, T.; Yu, Y. Molecular hydrogen protects mice against polymicrobial sepsis by ameliorating endothelial dysfunction via an Nrf2/HO-1 signaling pathway. Int. Immunopharmacol. 2015, 28, 643–654. [Google Scholar] [CrossRef]
  114. Ren, X.; Li, Y.; Zhou, Y.; Hu, W.; Yang, C.; Jing, Q.; Zhou, C.; Wang, X.; Hu, J.; Wang, L.; et al. Overcoming the compensatory elevation of NRF2 renders hepatocellular carcinoma cells more vulnerable to disulfiram/copper-induced ferroptosis. Redox Biol. 2021, 46, 102122. [Google Scholar] [CrossRef] [PubMed]
  115. La Rosa, P.; Petrillo, S.; Turchi, R.; Berardinelli, F.; Schirinzi, T.; Vasco, G.; Lettieri-Barbato, D.; Fiorenza, M.T.; Bertini, E.S.; Aquilano, K.; et al. The Nrf2 induction prevents ferroptosis in Friedreich’s Ataxia. Redox Biol. 2021, 38, 101791. [Google Scholar] [CrossRef] [PubMed]
  116. Wu, W.; Geng, P.; Zhu, J.; Li, J.; Zhang, L.; Chen, W.; Zhang, D.; Lu, Y.; Xu, X. KLF2 regulates eNOS uncoupling via Nrf2/HO-1 in endothelial cells under hypoxia and reoxygenation. Chem.-Biol. Interact. 2019, 305, 105–111. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, J.; Jiang, M.; Chen, X.; Montaner, L.J. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: Review of 3939 COVID-19 patients in China and emerging pathogenesis and therapy concepts. J. Leukoc. Biol. 2020, 108, 17–41. [Google Scholar] [CrossRef] [PubMed]
  118. Guo, R.F.; Ward, P.A. Role of oxidants in lung injury during sepsis. Antioxid. Redox Signal. 2007, 9, 1991–2002. [Google Scholar] [CrossRef] [PubMed]
  119. Holloway, P.M.; Gillespie, S.; Becker, F.; Vital, S.A.; Nguyen, V.; Alexander, J.S.; Evans, P.C.; Gavins, F.N.E. Sulforaphane induces neurovascular protection against a systemic inflammatory challenge via both Nrf2-dependent and independent pathways. Vasc. Pharmacol. 2016, 85, 29–38. [Google Scholar] [CrossRef] [Green Version]
  120. Kim, J.H.; Choi, Y.K.; Lee, K.S.; Cho, D.H.; Baek, Y.Y.; Lee, D.K.; Ha, K.S.; Choe, J.; Won, M.H.; Jeoung, D.; et al. Functional dissection of Nrf2-dependent phase II genes in vascular inflammation and endotoxic injury using Keap1 siRNA. Free Radic. Biol. Med. 2012, 53, 629–640. [Google Scholar] [CrossRef]
  121. Savla, S.R.; Prabhavalkar, K.S.; Bhatt, L.K. Cytokine storm associated coagulation complications in COVID-19 patients: Pathogenesis and Management. Expert Rev. Anti-Infect. Ther. 2021, 19, 1397–1413. [Google Scholar] [CrossRef]
  122. Terpos, E.; Ntanasis-Stathopoulos, I.; Elalamy, I.; Kastritis, E.; Sergentanis, T.N.; Politou, M.; Psaltopoulou, T.; Gerotziafas, G.; Dimopoulos, M.A. Hematological findings and complications of COVID-19. Am. J. Hematol. 2020, 95, 834–847. [Google Scholar] [CrossRef] [Green Version]
  123. Iba, T.; Connors, J.M.; Levy, J.H. The coagulopathy, endotheliopathy, and vasculitis of COVID-19. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2020, 69, 1181–1189. [Google Scholar] [CrossRef]
  124. Cao, T.H.; Jin, S.G.; Fei, D.S.; Kang, K.; Jiang, L.; Lian, Z.Y.; Pan, S.H.; Zhao, M.R.; Zhao, M.Y. Artesunate Protects Against Sepsis-Induced Lung Injury Via Heme Oxygenase-1 Modulation. Inflammation 2016, 39, 651–662. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, G.; Wu, Y.; Jin, S.; Sun, J.; Wan, B.B.; Zhang, J.; Wang, Y.; Gao, Z.Q.; Chen, D.; Li, S.; et al. Itaconate ameliorates methicillin-resistant Staphylococcus aureus-induced acute lung injury through the Nrf2/ARE pathway. Ann. Transl. Med. 2021, 9, 712. [Google Scholar] [CrossRef]
  126. Giustina, A.D.; Bonfante, S.; Zarbato, G.F.; Danielski, L.G.; Mathias, K.; de Oliveira, A.N., Jr.; Garbossa, L.; Cardoso, T.; Fileti, M.E.; De Carli, R.J.; et al. Dimethyl Fumarate Modulates Oxidative Stress and Inflammation in Organs after Sepsis in Rats. Inflammation 2018, 41, 315–327. [Google Scholar] [CrossRef] [PubMed]
  127. Thimmulappa, R.K.; Scollick, C.; Traore, K.; Yates, M.; Trush, M.A.; Liby, K.T.; Sporn, M.B.; Yamamoto, M.; Kensler, T.W.; Biswal, S. Nrf2-dependent protection from LPS induced inflammatory response and mortality by CDDO-Imidazolide. Biochem. Biophys. Res. Commun. 2006, 351, 883–889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Thimmulappa, R.K.; Fuchs, R.J.; Malhotra, D.; Scollick, C.; Traore, K.; Bream, J.H.; Trush, M.A.; Liby, K.T.; Sporn, M.B.; Kensler, T.W.; et al. Preclinical evaluation of targeting the Nrf2 pathway by triterpenoids (CDDO-Im and CDDO-Me) for protection from LPS-induced inflammatory response and reactive oxygen species in human peripheral blood mononuclear cells and neutrophils. Antioxid. Redox Signal. 2007, 9, 1963–1970. [Google Scholar] [CrossRef]
  129. Noel, S.; Zheng, L.; Navas-Acien, A.; Fuchs, R.J. The effect of ex vivo CDDO-Me activation on nuclear factor erythroid 2-related factor 2 pathway in white blood cells from patients with septic shock. Shock 2014, 42, 392–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Cai, Z.Y.; Sheng, Z.X.; Yao, H. Pachymic acid ameliorates sepsis-induced acute kidney injury by suppressing inflammation and activating the Nrf2/HO-1 pathway in rats. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1924–1931. [Google Scholar] [PubMed]
  131. Huang, X.T.; Liu, W.; Zhou, Y.; Hao, C.X.; Zhou, Y.; Zhang, C.Y.; Sun, C.C.; Luo, Z.Q.; Tang, S.Y. Dihydroartemisinin attenuates lipopolysaccharide-induced acute lung injury in mice by suppressing NF-kappaB signaling in an Nrf2-dependent manner. Int. J. Mol. Med. 2019, 44, 2213–2222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Meng, T.; Fu, S.; He, D.; Hu, G.; Gao, X.; Zhang, Y.; Huang, B.; Du, J.; Zhou, A.; Su, Y.; et al. Evodiamine Inhibits Lipopolysaccharide (LPS)-Induced Inflammation in BV-2 Cells via Regulating AKT/Nrf2-HO-1/NF-kappaB Signaling Axis. Cell. Mol. Neurobiol. 2021, 41, 115–127. [Google Scholar] [CrossRef] [PubMed]
  133. Li, S.T.; Dai, Q.; Zhang, S.X.; Liu, Y.J.; Yu, Q.Q.; Tan, F.; Lu, S.H.; Wang, Q.; Chen, J.W.; Huang, H.Q.; et al. Ulinastatin attenuates LPS-induced inflammation in mouse macrophage RAW264.7 cells by inhibiting the JNK/NF-kappaB signaling pathway and activating the PI3K/Akt/Nrf2 pathway. Acta Pharmacol. Sin. 2018, 39, 1294–1304. [Google Scholar] [CrossRef]
  134. Feng, Y.; Cui, R.; Li, Z.; Zhang, X.; Jia, Y.; Zhang, X.; Shi, J.; Qu, K.; Liu, C.; Zhang, J. Methane Alleviates Acetaminophen-Induced Liver Injury by Inhibiting Inflammation, Oxidative Stress, Endoplasmic Reticulum Stress, and Apoptosis through the Nrf2/HO-1/NQO1 Signaling Pathway. Oxid. Med. Cell. Longev. 2019, 2019, 7067619. [Google Scholar] [CrossRef] [PubMed]
  135. Motterlini, R.; Nikam, A.; Manin, S.; Ollivier, A.; Wilson, J.L.; Djouadi, S.; Muchova, L.; Martens, T.; Rivard, M.; Foresti, R. HYCO-3, a dual CO-releaser/Nrf2 activator, reduces tissue inflammation in mice challenged with lipopolysaccharide. Redox Biol. 2019, 20, 334–348. [Google Scholar] [CrossRef]
  136. Wruck, C.J.; Streetz, K.; Pavic, G.; Gotz, M.E.; Tohidnezhad, M.; Brandenburg, L.O.; Varoga, D.; Eickelberg, O.; Herdegen, T.; Trautwein, C.; et al. Nrf2 induces interleukin-6 (IL-6) expression via an antioxidant response element within the IL-6 promoter. J. Biol. Chem. 2011, 286, 4493–4499. [Google Scholar] [CrossRef] [Green Version]
  137. Lv, P.; Xue, P.; Dong, J.; Peng, H.; Clewell, R.; Wang, A.; Wang, Y.; Peng, S.; Qu, W.; Zhang, Q.; et al. Keap1 silencing boosts lipopolysaccharide-induced transcription of interleukin 6 via activation of nuclear factor kappaB in macrophages. Toxicol. Appl. Pharmacol. 2013, 272, 697–702. [Google Scholar] [CrossRef]
  138. Wang, Y.; Wang, B.; Du, F.; Su, X.; Sun, G.; Zhou, G.; Bian, X.; Liu, N. Epigallocatechin-3-Gallate Attenuates Oxidative Stress and Inflammation in Obstructive Nephropathy via NF-kappaB and Nrf2/HO-1 Signalling Pathway Regulation. Basic Clin. Pharmacol. Toxicol. 2015, 117, 164–172. [Google Scholar] [CrossRef]
  139. Pedruzzi, L.M.; Stockler-Pinto, M.B.; Leite, M., Jr.; Mafra, D. Nrf2-keap1 system versus NF-kappaB: The good and the evil in chronic kidney disease? Biochimie 2012, 94, 2461–2466. [Google Scholar] [CrossRef]
  140. Park, S.Y.; Jin, M.L.; Wang, Z.; Park, G.; Choi, Y.W. 2,3,4’,5-tetrahydroxystilbene-2-O-beta-d-glucoside exerts anti-inflammatory effects on lipopolysaccharide-stimulated microglia by inhibiting NF-kappaB and activating AMPK/Nrf2 pathways. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2016, 97, 159–167. [Google Scholar] [CrossRef]
  141. Tan, Y.; Wan, H.H.; Sun, M.M.; Zhang, W.J.; Dong, M.; Ge, W.; Ren, J.; Peng, H. Cardamonin protects against lipopolysaccharide-induced myocardial contractile dysfunction in mice through Nrf2-regulated mechanism. Acta Pharmacol. Sin. 2021, 42, 404–413. [Google Scholar] [CrossRef]
  142. Zhong, W.; Qian, K.; Xiong, J.; Ma, K.; Wang, A.; Zou, Y. Curcumin alleviates lipopolysaccharide induced sepsis and liver failure by suppression of oxidative stress-related inflammation via PI3K/AKT and NF-kappaB related signaling. Biomed. Pharmacother. Biomed. Pharmacother. 2016, 83, 302–313. [Google Scholar] [CrossRef] [PubMed]
  143. Dai, J.P.; Wang, Q.W.; Su, Y.; Gu, L.M.; Zhao, Y.; Chen, X.X.; Chen, C.; Li, W.Z.; Wang, G.F.; Li, K.S. Emodin Inhibition of Influenza A Virus Replication and Influenza Viral Pneumonia via the Nrf2, TLR4, p38/JNK and NF-kappaB Pathways. Molecules 2017, 22, 1754. [Google Scholar] [CrossRef] [PubMed]
  144. Pei, X.; Zhang, X.J.; Chen, H.M. Bardoxolone treatment alleviates lipopolysaccharide (LPS)-induced acute lung injury through suppressing inflammation and oxidative stress regulated by Nrf2 signaling. Biochem. Biophys. Res. Commun. 2019, 516, 270–277. [Google Scholar] [CrossRef] [PubMed]
  145. Son, Y.; Kim, S.; Chung, H.T.; Pae, H.O. Reactive oxygen species in the activation of MAP kinases. Methods Enzymol. 2013, 528, 27–48. [Google Scholar] [CrossRef] [PubMed]
  146. Arthur, J.S.; Ley, S.C. Mitogen-activated protein kinases in innate immunity. Nat. Rev. Immunol. 2013, 13, 679–692. [Google Scholar] [CrossRef]
  147. Dai, J.; Gu, L.; Su, Y.; Wang, Q.; Zhao, Y.; Chen, X.; Deng, H.; Li, W.; Wang, G.; Li, K. Inhibition of curcumin on influenza A virus infection and influenzal pneumonia via oxidative stress, TLR2/4, p38/JNK MAPK and NF-kappaB pathways. Int. Immunopharmacol. 2018, 54, 177–187. [Google Scholar] [CrossRef]
  148. Kim, T.H.; Yoon, S.J.; Lee, S.M. Genipin attenuates sepsis by inhibiting Toll-like receptor signaling. Mol. Med. 2012, 18, 455–465. [Google Scholar] [CrossRef]
  149. Lu, H.; Wang, B.; Cui, N.; Zhang, Y. Artesunate suppresses oxidative and inflammatory processes by activating Nrf2 and ROS-dependent p38 MAPK and protects against cerebral ischemia-reperfusion injury. Mol. Med. Rep. 2018, 17, 6639–6646. [Google Scholar] [CrossRef] [Green Version]
  150. Hou, Y.; Xue, P.; Bai, Y.; Liu, D.; Woods, C.G.; Yarborough, K.; Fu, J.; Zhang, Q.; Sun, G.; Collins, S.; et al. Nuclear factor erythroid-derived factor 2-related factor 2 regulates transcription of CCAAT/enhancer-binding protein beta during adipogenesis. Free Radic. Biol. Med. 2012, 52, 462–472. [Google Scholar] [CrossRef] [Green Version]
  151. Pi, J.; Leung, L.; Xue, P.; Wang, W.; Hou, Y.; Liu, D.; Yehuda-Shnaidman, E.; Lee, C.; Lau, J.; Kurtz, T.W.; et al. Deficiency in the nuclear factor E2-related factor-2 transcription factor results in impaired adipogenesis and protects against diet-induced obesity. J. Biol. Chem. 2010, 285, 9292–9300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Li, L.; Fu, J.; Liu, D.; Sun, J.; Hou, Y.; Chen, C.; Shao, J.; Wang, L.; Wang, X.; Zhao, R.; et al. Hepatocyte-specific Nrf2 deficiency mitigates high-fat diet-induced hepatic steatosis: Involvement of reduced PPARgamma expression. Redox Biol. 2020, 30, 101412. [Google Scholar] [CrossRef]
  153. Cho, H.Y.; Gladwell, W.; Wang, X.; Chorley, B.; Bell, D.; Reddy, S.P.; Kleeberger, S.R. Nrf2-regulated PPARgamma expression is critical to protection against acute lung injury in mice. Am. J. Respir. Crit. Care Med. 2010, 182, 170–182. [Google Scholar] [CrossRef] [Green Version]
  154. Hovsepian, E.; Penas, F.; Goren, N.B. 15-deoxy-Delta12,14 prostaglandin GJ2 but not rosiglitazone regulates metalloproteinase 9, NOS-2, and cyclooxygenase 2 expression and functions by peroxisome proliferator-activated receptor gamma-dependent and -independent mechanisms in cardiac cells. Shock 2010, 34, 60–67. [Google Scholar] [CrossRef]
  155. von Knethen, A.; Soller, M.; Brune, B. Peroxisome proliferator-activated receptor gamma (PPAR gamma) and sepsis. Arch. Immunol. Ther. Exp. 2007, 55, 19–25. [Google Scholar] [CrossRef]
  156. Ciavarella, C.; Motta, I.; Valente, S.; Pasquinelli, G. Pharmacological (or Synthetic) and Nutritional Agonists of PPAR-gamma as Candidates for Cytokine Storm Modulation in COVID-19 Disease. Molecules 2020, 25, 2076. [Google Scholar] [CrossRef] [PubMed]
  157. Mansour, H.H.; El Kiki, S.M.; Galal, S.M. Metformin and low dose radiation modulates cisplatin-induced oxidative injury in rat via PPAR-gamma and MAPK pathways. Arch. Biochem. Biophys. 2017, 616, 13–19. [Google Scholar] [CrossRef] [PubMed]
  158. Tian, H.; Zhang, B.; Di, J.; Jiang, G.; Chen, F.; Li, H.; Li, L.; Pei, D.; Zheng, J. Keap1: One stone kills three birds Nrf2, IKKbeta and Bcl-2/Bcl-xL. Int. J. Mol. Sci. 2012, 325, 26–34. [Google Scholar] [CrossRef]
  159. Lee, D.F.; Kuo, H.P.; Liu, M.; Chou, C.K.; Xia, W.; Du, Y.; Shen, J.; Chen, C.T.; Huo, L.; Hsu, M.C.; et al. KEAP1 E3 ligase-mediated downregulation of NF-kappaB signaling by targeting IKKbeta. Mol. Cell 2009, 36, 131–140. [Google Scholar] [CrossRef] [Green Version]
  160. Kim, J.E.; You, D.J.; Lee, C.; Ahn, C.; Seong, J.Y.; Hwang, J.I. Suppression of NF-kappaB signaling by KEAP1 regulation of IKKbeta activity through autophagic degradation and inhibition of phosphorylation. Cell. Signal. 2010, 22, 1645–1654. [Google Scholar] [CrossRef]
  161. Zhao, R.; Hou, Y.; Zhang, Q.; Woods, C.G.; Xue, P.; Fu, J.; Yarborough, K.; Guan, D.; Andersen, M.E.; Pi, J. Cross-regulations among NRFs and KEAP1 and effects of their silencing on arsenic-induced antioxidant response and cytotoxicity in human keratinocytes. Environ. Health Perspect. 2012, 120, 583–589. [Google Scholar] [CrossRef] [Green Version]
  162. Gong, H.; Tai, H.; Huang, N.; Xiao, P.; Mo, C.; Wang, X.; Han, X.; Zhou, J.; Chen, H.; Tang, X.; et al. Nrf2-SHP Cascade-Mediated STAT3 Inactivation Contributes to AMPK-Driven Protection Against Endotoxic Inflammation. Front. Immunol. 2020, 11, 414. [Google Scholar] [CrossRef] [Green Version]
  163. Yuk, J.M.; Jin, H.S.; Jo, E.K. Small Heterodimer Partner and Innate Immune Regulation. Endocrinol. Metab. 2016, 31, 17–24. [Google Scholar] [CrossRef]
  164. Gawish, R.A.; Fahmy, H.A.; Abd El Fattah, A.I.; Nada, A.S. The potential effect of methylseleninic acid (MSA) against gamma-irradiation induced testicular damage in rats: Impact on JAK/STAT pathway. Arch. Biochem. Biophys. 2020, 679, 108205. [Google Scholar] [CrossRef]
  165. Netea, M.G.; van de Veerdonk, F.L.; van der Meer, J.W.; Dinarello, C.A.; Joosten, L.A. Inflammasome-independent regulation of IL-1-family cytokines. Annu. Rev. Immunol. 2015, 33, 49–77. [Google Scholar] [CrossRef]
  166. Rahim, I.; Sayed, R.K.; Fernandez-Ortiz, M.; Aranda-Martinez, P.; Guerra-Librero, A.; Fernandez-Martinez, J.; Rusanova, I.; Escames, G.; Djerdjouri, B.; Acuna-Castroviejo, D. Melatonin alleviates sepsis-induced heart injury through activating the Nrf2 pathway and inhibiting the NLRP3 inflammasome. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2021, 394, 261–277. [Google Scholar] [CrossRef] [PubMed]
  167. Xie, K.; Zhang, Y.; Wang, Y.; Meng, X.; Wang, Y.; Yu, Y.; Chen, H. Hydrogen attenuates sepsis-associated encephalopathy by NRF2 mediated NLRP3 pathway inactivation. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2020, 69, 697–710. [Google Scholar] [CrossRef] [PubMed]
  168. Olagnier, D.; Farahani, E.; Thyrsted, J.; Blay-Cadanet, J.; Herengt, A.; Idorn, M.; Hait, A.; Hernaez, B.; Knudsen, A.; Iversen, M.B.; et al. SARS-CoV2-mediated suppression of NRF2-signaling reveals potent antiviral and anti-inflammatory activity of 4-octyl-itaconate and dimethyl fumarate. Nat. Commun. 2020, 11, 4938. [Google Scholar] [CrossRef] [PubMed]
  169. Desai, N.; Neyaz, A.; Szabolcs, A.; Shih, A.R.; Chen, J.H.; Thapar, V.; Nieman, L.T.; Solovyov, A.; Mehta, A.; Lieb, D.J.; et al. Temporal and spatial heterogeneity of host response to SARS-CoV-2 pulmonary infection. Nat. Commun. 2020, 11, 6319. [Google Scholar] [CrossRef] [PubMed]
  170. Robledinos-Anton, N.; Fernandez-Gines, R.; Manda, G.; Cuadrado, A. Activators and Inhibitors of NRF2: A Review of Their Potential for Clinical Development. Oxid. Med. Cell. Longev. 2019, 2019, 9372182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Zarbato, G.F.; de Souza Goldim, M.P.; Giustina, A.D.; Danielski, L.G.; Mathias, K.; Florentino, D.; de Oliveira Junior, A.N.; da Rosa, N.; Laurentino, A.O.; Trombetta, T.; et al. Dimethyl Fumarate Limits Neuroinflammation and Oxidative Stress and Improves Cognitive Impairment After Polymicrobial Sepsis. Neurotox. Res. 2018, 34, 418–430. [Google Scholar] [CrossRef]
  172. Qi, T.; Xu, F.; Yan, X.; Li, S.; Li, H. Sulforaphane exerts anti-inflammatory effects against lipopolysaccharide-induced acute lung injury in mice through the Nrf2/ARE pathway. Int. J. Mol. Med. 2016, 37, 182–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Lee, I.C.; Kim, D.Y.; Bae, J.S. Sulforaphane Reduces HMGB1-Mediated Septic Responses and Improves Survival Rate in Septic Mice. Am. J. Chin. Med. 2017, 45, 1253–1271. [Google Scholar] [CrossRef] [PubMed]
  174. Ku, S.K.; Han, M.S.; Bae, J.S. Sulforaphane inhibits endothelial protein C receptor shedding in vitro and in vivo. Vasc. Pharmacol. 2014, 63, 13–18. [Google Scholar] [CrossRef]
  175. Xu, Y.; Liu, L. Curcumin alleviates macrophage activation and lung inflammation induced by influenza virus infection through inhibiting the NF-kappaB signaling pathway. Influ. Respir. Viruses 2017, 11, 457–463. [Google Scholar] [CrossRef] [PubMed]
  176. Valizadeh, H.; Abdolmohammadi-Vahid, S.; Danshina, S.; Ziya Gencer, M.; Ammari, A.; Sadeghi, A.; Roshangar, L.; Aslani, S.; Esmaeilzadeh, A.; Ghaebi, M.; et al. Nano-curcumin therapy, a promising method in modulating inflammatory cytokines in COVID-19 patients. Int. Immunopharmacol. 2020, 89, 107088. [Google Scholar] [CrossRef]
  177. Ali, F.E.M.; Bakr, A.G.; Abo-Youssef, A.M.; Azouz, A.A.; Hemeida, R.A.M. Targeting Keap-1/Nrf-2 pathway and cytoglobin as a potential protective mechanism of diosmin and pentoxifylline against cholestatic liver cirrhosis. Life Sci. 2018, 207, 50–60. [Google Scholar] [CrossRef]
  178. Patangrao Renushe, A.; Kumar Banothu, A.; Kumar Bharani, K.; Mekala, L.; Mahesh Kumar, J.; Neeradi, D.; Durga Veera Hanuman, D.; Gadige, A.; Khurana, A. Vincamine, an active constituent of Vinca rosea ameliorates experimentally induced acute lung injury in Swiss albino mice through modulation of Nrf-2/NF-kappaB signaling cascade. Int. Immunopharmacol. 2022, 108, 108773. [Google Scholar] [CrossRef]
  179. Khurana, A.; Sikha, M.S.; Ramesh, K.; Venkatesh, P.; Godugu, C. Modulation of cerulein-induced pancreatic inflammation by hydroalcoholic extract of curry leaf (Murraya koenigii). Phytother. Res. PTR 2019, 33, 1510–1525. [Google Scholar] [CrossRef] [PubMed]
  180. Collins, S.; Pi, J.; Yehuda-Shnaidman, E. Uncoupling and reactive oxygen species (ROS)—A double-edged sword for beta-cell function? “Moderation in all things”. Best Pract. Res. Clin. Endocrinol. Metab. 2012, 26, 753–758. [Google Scholar] [CrossRef] [PubMed]
  181. Pi, J.; Zhang, Q.; Fu, J.; Woods, C.G.; Hou, Y.; Corkey, B.E.; Collins, S.; Andersen, M.E. ROS signaling, oxidative stress and Nrf2 in pancreatic beta-cell function. Toxicol. Appl. Pharmacol. 2010, 244, 77–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Hu, Y.; Li, J.; Lou, B.; Wu, R.; Wang, G.; Lu, C.; Wang, H.; Pi, J.; Xu, Y. The Role of Reactive Oxygen Species in Arsenic Toxicity. Biomolecules 2020, 10, 240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Li, C.; Yan, Y.; Shi, Q.; Kong, Y.; Gao, L.; Bao, H.; Li, Y. Recuperating lung decoction attenuates inflammation and oxidation in cigarette smoke-induced COPD in rats via activation of ERK and Nrf2 pathways. Cell Biochem. Funct. 2017, 35, 278–286. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of pathogen-induced cytokine storms. The PAMPs and DAMPs derived from PAMPs–induced cell damages are the primary initiators of cytokine storms, in which multiple key cellular events are involved. Without proper antioxidant and/or anti-inflammatory response, accumulating ROS and cytokines may form a positive feedback loop to aggravate inflammatory response at the infection loci, leading to a pathological burst of pro-inflammatory cytokines into the systemic circulation and multi-organ failure.
Figure 1. Schematic illustration of pathogen-induced cytokine storms. The PAMPs and DAMPs derived from PAMPs–induced cell damages are the primary initiators of cytokine storms, in which multiple key cellular events are involved. Without proper antioxidant and/or anti-inflammatory response, accumulating ROS and cytokines may form a positive feedback loop to aggravate inflammatory response at the infection loci, leading to a pathological burst of pro-inflammatory cytokines into the systemic circulation and multi-organ failure.
Antioxidants 12 00172 g001
Figure 2. The vicious cycle between ROS and cytokines in cytokine storms. The antioxidant system negatively regulates intracellular ROS levels, which in turn affect the activity of the antioxidant system via negative feedback mechanisms to maintain redox homeostasis. Uncontrolled production of ROS may activate multiple inflammatory signaling cascades, including NF-κB and MAPK, to produce cytokines, which may in turn stimulate mitochondria and NOX family to increase ROS production. Without proper antioxidant and/or anti-inflammatory responses, the overproduced ROS and cytokines may form a positive feedback loop to amplify the inflammatory response, leading to cytokine storms.
Figure 2. The vicious cycle between ROS and cytokines in cytokine storms. The antioxidant system negatively regulates intracellular ROS levels, which in turn affect the activity of the antioxidant system via negative feedback mechanisms to maintain redox homeostasis. Uncontrolled production of ROS may activate multiple inflammatory signaling cascades, including NF-κB and MAPK, to produce cytokines, which may in turn stimulate mitochondria and NOX family to increase ROS production. Without proper antioxidant and/or anti-inflammatory responses, the overproduced ROS and cytokines may form a positive feedback loop to amplify the inflammatory response, leading to cytokine storms.
Antioxidants 12 00172 g002
Figure 3. NRF2 modulates inflammatory response at different levels via multiple cellular events. (1) Macrophage polarization: NRF2-mediated antioxidant response can suppress NF-κB and MAPK to prevent macrophage polarization toward M1. In addition, NRF2-dependent induction of HO-1 can promote M2 polarization and thus promote anti-inflammatory response; (2) Pyroptosis: NRF2-mediated induction of antioxidants can decrease the accumulation of intracellular ROS and subsequently caspase-1 and 3 activation and thus pyroptosis; (3) Endothelial dysfunction: NRF2-dependent expression of antioxidant enzymes is crucial in controlling the production and secretion of NO and chemokines, and thus protecting endothelial cells from malfunction and various RCD; (4) Neutrophil infiltration: NRF2-mediated antioxidant response can suppress neutrophil recruitment and neutrophil extracellular traps (NETs) formation, resulting in reduced secretion of inflammatory cytokines.
Figure 3. NRF2 modulates inflammatory response at different levels via multiple cellular events. (1) Macrophage polarization: NRF2-mediated antioxidant response can suppress NF-κB and MAPK to prevent macrophage polarization toward M1. In addition, NRF2-dependent induction of HO-1 can promote M2 polarization and thus promote anti-inflammatory response; (2) Pyroptosis: NRF2-mediated induction of antioxidants can decrease the accumulation of intracellular ROS and subsequently caspase-1 and 3 activation and thus pyroptosis; (3) Endothelial dysfunction: NRF2-dependent expression of antioxidant enzymes is crucial in controlling the production and secretion of NO and chemokines, and thus protecting endothelial cells from malfunction and various RCD; (4) Neutrophil infiltration: NRF2-mediated antioxidant response can suppress neutrophil recruitment and neutrophil extracellular traps (NETs) formation, resulting in reduced secretion of inflammatory cytokines.
Antioxidants 12 00172 g003
Figure 4. Molecular details underlying NRF2-mediated downregulation of cytokines. The cytokine production process includes transcription, translation, and maturation. NF-κB, STAT3, and MAPK-activated transcription factors can promote the transcriptional process. By induction of antioxidant enzymes, NRF2 can suppress NF-κB and MAPK in an ROS-dependent manner. In addition, NRF2 can also inhibit the activation of NF-κB and STAT3 via induction of PPAR-γ and small heterodimer protein (SHP), respectively. In addition, NRF2 can suppress NLRP3 inflammasome and thus IL-1β maturation in an antioxidant-dependent mechanism.
Figure 4. Molecular details underlying NRF2-mediated downregulation of cytokines. The cytokine production process includes transcription, translation, and maturation. NF-κB, STAT3, and MAPK-activated transcription factors can promote the transcriptional process. By induction of antioxidant enzymes, NRF2 can suppress NF-κB and MAPK in an ROS-dependent manner. In addition, NRF2 can also inhibit the activation of NF-κB and STAT3 via induction of PPAR-γ and small heterodimer protein (SHP), respectively. In addition, NRF2 can suppress NLRP3 inflammasome and thus IL-1β maturation in an antioxidant-dependent mechanism.
Antioxidants 12 00172 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liu, Z.; Deng, P.; Liu, S.; Bian, Y.; Xu, Y.; Zhang, Q.; Wang, H.; Pi, J. Is Nuclear Factor Erythroid 2-Related Factor 2 a Target for the Intervention of Cytokine Storms? Antioxidants 2023, 12, 172. https://doi.org/10.3390/antiox12010172

AMA Style

Liu Z, Deng P, Liu S, Bian Y, Xu Y, Zhang Q, Wang H, Pi J. Is Nuclear Factor Erythroid 2-Related Factor 2 a Target for the Intervention of Cytokine Storms? Antioxidants. 2023; 12(1):172. https://doi.org/10.3390/antiox12010172

Chicago/Turabian Style

Liu, Zihang, Panpan Deng, Shengnan Liu, Yiying Bian, Yuanyuan Xu, Qiang Zhang, Huihui Wang, and Jingbo Pi. 2023. "Is Nuclear Factor Erythroid 2-Related Factor 2 a Target for the Intervention of Cytokine Storms?" Antioxidants 12, no. 1: 172. https://doi.org/10.3390/antiox12010172

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