The NRF2 Pathway Regulates Cellular Responses to Stress: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , , ,

NRF2 belongs to the cap “n” collar (CNC) family of transcription factors and is found in the cytoplasm of non-stressed cells in a combined form with KEAP1. Oxidative stress activates the transcription factor NRF2, which plays a key role in alleviating redox-induced cellular injury.

  • NRF2 pathway
  • respiratory viruses
  • viral replication
  • inflammation

1. Introduction

Respiratory viruses target the human respiratory system and cause various clinical symptoms in humans, ranging from mild upper respiratory infections to organ failure and life-threatening respiratory diseases [1,2]. The most common respiratory viruses are rhinoviruses, coronaviruses (CoVs), influenza virus, respiratory syncytial virus (RSV), parainfluenza viruses, enteroviruses, adenoviruses, and human metapneumovirus (hMPV) [3]. Each year, nearly 4 million deaths are attributed to lower respiratory tract infections, with Influenza contributing to approximately half a million of these fatalities [4]. Moreover, morbidity and mortality caused by respiratory viruses increased drastically with the emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the COVID-19 pandemic. Although several host factors have been found to play crucial role in the pathogenesis of respiratory viral infections, the interaction between respiratory viruses and the host cellular response remains poorly understood. Understanding antiviral host pathways and defining their role in the pathogenesis of respiratory viruses may set the foundation for novel antiviral therapies for viral respiratory diseases.
Viral respiratory infections are commonly associated with increased production of reactive oxygen and nitrogen species (ROS and RNS), leading to oxidative stress [5,6]. Subsequently, increased oxidative stress contributes to reduced host antiviral response, enhanced replication and virus-induced cell and tissue injury apoptosis, ferroptosis, inflammation, causing organ damage [6,7] and the occurrence of clinical symptoms [5,8,9]. During oxidative stress induced by respiratory viruses, the host deploys antioxidant mechanisms to control signaling pathways and reestablish cellular redox balance. Many respiratory viruses, including influenza [10,11,12,13,14,15,16,17], CoVs [18,19,20,21,22,23,24,25,26], RSV [27,28,29,30,31,32,33,34,35,36,37,38,39,40], rhinoviruses [41,42,43,44,45], enteroviruses such as Coxsackievirus B3 (CVB3) [46], EV71 [47,48,49,50], metapneumoviruses [34], and parainfluenza viruses [51,52], have been demonstrated to disrupt the cell redox homeostasis and induce the production of ROS.
However, in response to virus-induced oxidative stress, host cells deploy a strong antioxidant response characterized by the production of proteins (enzymes) and/or small molecules (vitamins C and E) that are mainly mediated by the nuclear factor erythroid 2-related factor (NRF2) to counteract the redox-induced toxicity and restore cellular redox homeostasis [53,54].

2. The NRF2 Pathway Regulates Cellular Responses to Stress

The production of ROS and activation of an antioxidant response is known to be controlled by the Kelch-like ECH-associated protein 1 (KEAP1)–NRF2 axis. This regulation occurs through intrinsic mechanisms within different cell types of the airway epithelium (e.g., nasal versus bronchial cells). NRF2 belongs to the cap “n” collar (CNC) family of transcription factors and is found in the cytoplasm of non-stressed cells in a combined form with KEAP1. In quiescent cells, an adapter protein, KEAP1, interacts with NRF2 and recruits cullin-3 (CUL3)-containing E3 ubiquitin ligase to form a complex that regulates the ubiquitination of NRF2. Consequently, polyubiquitination of NRF2 leads to NRF2 degradation via the 26S proteasome machinery, which ensures that the NRF2 level and its activity remain low during redox homeostasis [55]. Contrarily, during a viral respiratory infection or other induced oxidative stress, NRF2 escapes repression by KEAP1. The CUL3/KEAP1 complex that targets NRF2 for ubiquitination undergoes a change to a nonfunctional conformation [56,57,58,59]. Thus, upon activation, newly synthesized NRF2 is no longer ubiquitinated/degraded, rapidly accumulates, and translocates to the nucleus where it binds the small maf protein (sMaf) [56,57,58,59]. The NRF2–Maf heterodimer binds to the antioxidant response element (ARE) (or multiple Maf recognition elements (MAREs)). This interaction induces the transcription of a wide variety of antioxidant genes, including HO-1 and genes that are involved in the synthesis and recycling of glutathione (Figure 1). A heme sensor known as BTB and CNC homology 1 (BACH-1) can also bind to the ARE in a KEAP1-independent manner and directly competes with NRF2 for binding to AREs. The interaction of BACH-1 with ARE prevents NRF2 from binding to the ARE, thus repressing HO-1 [60,61,62,63,64,65] (Figure 1). HO-1 catalyzes the degradation of heme into carbon monoxide (CO), Fe2+, and biliverdin, and has antiviral properties through multiple pathways (Figure 2). Importantly, NRF2 activation appears to have an inhibitory effect on the interferon response, which is an important component of the innate immune system’s antiviral defense. The balance between NRF2 activation and the interferon response is regulated by intrinsic cell factors, for which the cell composition varies from nasal to bronchial cells, and potentially influences the susceptibility to viral infections. Notably, activation of the NRF2 pathway appears to mediate protection against viral respiratory infections, including SARS-CoV-2, influenza viruses, and several RNA and DNA viruses that induce oxidative stress (Supplementary Table S1) [5,8,9,10,11,12,13,14,18,19,20,21,22,23,24,27,28,29,30,31,32,33,34,35,36,41,42,43,44,46,47,48,49,51,52,53,54,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93]. Overall, the crosstalk between NRF2 and viruses is bidirectional and complex. With regards to the impact of viruses on the NRF2 pathway, respiratory viruses such as influenza [12,94], SARS-CoV-2 [19,20,22,24,95,96], RSV [27,31,32], rhinovirus [43,44], and enteroviruses [47] directly alter NRF2 levels and signaling [5,8,9,10,18,33] (Table 1). With regards to the impact of the NRF2 pathway on viruses, host NRF2 pathways also regulate the replication of several respiratory viruses such as influenza [11,12,13], coronaviruses [19,20,21,22,23], RSV [34,35], rhinovirus [44], enterovirus [47], metapneumovirus [34], and parainfluenza [51,52] (Table 2). Notably, the host NRF2 pathways also regulate viral replication, apoptosis, ferroptosis, and inflammation (Table 2, Table 3, Table 4, Table 5 and Table 6). Besides playing an essential role in cell defense against redox stresses by trans-activating cytoprotective genes encoding antioxidant and detoxifying enzymes, NRF2 contributes to the regulation of the anti-inflammatory response and metabolic reprogramming [97].
Figure 1. The nuclear factor erythroid 2-related factor 2 (NRF2) pathway regulates cellular responses to stress. (Left) Under resting (constitutive) condition, in the cytoplasm NRF2 is anchored with Kelch-like ECH-associated protein 1 (KEAP1). NRF2 binds KEAP1 and becomes ubiquinated, leading to degradation by the 26S proteasome. (Right) Under oxidative stress response, NRF2 escapes repression by KEAP1. The CUL3/KEAP1 complex that targets NRF2 for ubiquitination undergoes a change to a nonfunctional conformation. Thus, newly synthesized NRF2 is no longer ubiquitinated/degraded, rapidly accumulates, and translocates to the nucleus where it binds the small maf protein (sMaf) and antioxidant response element (ARE). Activation of ARE increases the expression of the antioxidant genes heme oxygenase 1 (HO-1), quinone oxidoreductase (NQO1), and glutathione (GSH), which blocks the progression of oxidative stress (OS). Thus, activation of the NRF2 pathway has cytoprotective effects and plays a key role in maintaining redox balance. Figure generated with Biorender (https://biorender.com/, accessed on 10 November 2023).
Figure 2. Cytoprotective effects of heme oxygenase 1 (HO-1), a key gene of the nuclear factor erythroid 2-related factor 2 (NRF2) pathway, in viral infection. HO-1 is a metabolic enzyme that utilizes oxygen (O2), heme, and NADPH to catalyze the degradation of heme into carbon monoxide (CO), Fe2+, and biliverdin. HO-1 has antiviral properties through multiple pathways: (1) Free Fe2+ may act on viral replication by binding to the highly conserved divalent metal-binding pocket of the viral RNA and inhibiting enzymes that mediate viral replication. (2) Biliverdin may inhibit viral proteases. (3) Heterodimerization of HO-1 with interferon regulatory factor 3 (IRF3) facilitates the phosphorylation and nuclear translocation of IRF3 and the induction of type I interferon (IFN) gene expression that has antiviral properties. (4) CO activates protein kinase G (PKG), which inhibits NAPDH oxidases (NOX), preventing an increase in reactive oxygen species (ROS) and associated damage. (5) Biliverdin also has antioxidant properties, and it is converted by NRF2/ARE-regulated gene biliverdin reductase to the potent antioxidant bilirubin. Figure generated with Biorender (https://biorender.com/, accessed on 10 November 2023).
Table 1. Impact of respiratory viruses on the NRF2 pathway.
Table 2. Impact of NRF2 on the replication of respiratory viruses.
Table 3. Antiviral activity of Heme Oxygenase 1 (HO-1), a key downstream gene of the NRF2 pathway.
Table 4. Respiratory viruses and the role of NRF2 role in inflammation.
Table 5. Impact of viruses and NRF2 on the apoptosis pathway.
Table 6. Impact of viruses and NRF2 on the ferroptosis pathway.
Viruses/NRF2 Impact on Ferroptosis Reference
NRF2 ∙ NRF2 ↓ ROS and ↑ antioxidant responses, and ↑ GPX4-induced ↓ of ferroptosis.
∙ NRF2 ↑ Heme Oxygenase 1 (HO-1) that ↓ ferroptosis.
∙ NRF2 ↑ antioxidant enzymes.
[9]
Influenza ∙ Iron ↓ viral genome amplification and viral replication.
∙ Influenza ↓ cellular GSH and/or affects GPX4 activity.
∙ Neuraminidase of Influenza A binds lysosome-associated membrane proteins and ↑ lysosome rupture.
[9,108,109,110,111,112]
SARS-CoV-2, SARS-CoV, other coronaviruses ∙ SARS-CoV-2 Potentially causes cellular iron overload and iron scavenging.
∙ SARS-CoV-2 ↑serum ferritin concentration.
∙ CoVs↓ cellular GSH and/or affect GPX4 activity.
∙ SARS-CoV ORF-3a viral protein ↑ lysosomal damage and dysfunction.
[113,114,115,116,117]
RSV ∙ ↑ the expression of 12/15-LOX and mitochondrial iron content. [118]
EV-71, CB3 ∙ Iron ↓ viral genome amplification and viral replication of EV-71.
∙ CB3 ↑the expression NRAMP (DMT) and ↑cellular iron uptake.
[108,119,120]
Non-respiratory viruses:
HBV, HCV, WNV, Dengue virus, HSV, KSHV
∙ HBV, HCV: ↑ serum and cellular iron uptake and ↓ hepcidin expression, ↑ serum ferritin concentration, and uses TfR1 as a cellular receptor.
∙ HIV ↓ serum iron, ↑ the expression of hepcidin, ↑cellular iron via hepcidin mediated degradation of ferroportin, ↓ cellular GSH and/or affects GPX4 activity, and upregulates the expression of system xc-.
∙ WNV ↑ the expression NRAMP (DMT) and ↑cellular iron uptake.
∙ Dengue virus ↓ cellular GSH and/or affects GPX4 activity.
∙ HSV ↓ cellular GSH and/or affects GPX4 activity.
∙ JEV ↓ cellular GSH and/or affects GPX4 activity, produces lipid peroxide free radicals, and ↑ the expression of system xc-.
∙ KSHV ↓ cellular GSH and/or affects GPX4 activity.
∙ Zika virus ↓ cellular GSH and/or affects GPX4 activity.
∙ Other viruses (e.g., hemorrhagic viruses) use NRAMP or TfR1 as a cellular receptor.
[121]
Abbreviations: CB3: Coxsackievirus B3, EV-71: Enterovirus 71, GSH: Glutathione, GPX4: Glutathione Peroxidase 4, HBV: Hepatitis B virus, HCV: Hepatitis C virus, HIV: human immunodeficiency virus, HSV: Herpes simplex virus, JEV: Japanese encephalitis virus, KSHV: Kaposi sarcoma-associated herpesvirus, 12/15-LOX:12/15-lipoxygenase, NRAMP: Natural Resistance-Associated Macrophage Proteins, ORF-3a: Open Reading Frame-3a, RSV: respiratory syncytial virus, SARS-CoV-2: severe acute respiratory syndrome-coronavirus-2, SARS-CoV: severe acute respiratory syndrome coronavirus, TfR1: transferrin receptor 1 protein, WNV: West Nile virus.

This entry is adapted from the peer-reviewed paper 10.3390/pathogens13010039

This entry is offline, you can click here to edit this entry!
Video Production Service