Back to Journals » Journal of Inflammation Research » Volume 17

Acute Lung Injury and the NLRP3 Inflammasome

Authors Gu W, Zeng Q, Wang X, Jasem H, Ma L

Received 20 February 2024

Accepted for publication 22 May 2024

Published 13 June 2024 Volume 2024:17 Pages 3801—3813

DOI https://doi.org/10.2147/JIR.S464838

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Ning Quan



Wanjun Gu, Qi Zeng, Xin Wang, Huthaifa Jasem, Ling Ma

Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China

Correspondence: Ling Ma, No. 36 Sanhao Street, Heping District, Shenyang, 110004, People’s Republic of China, Tel +86-18940256927, Email [email protected]

Abstract: Acute lung injury (ALI) manifests through harm to the capillary endothelium and alveolar epithelial cells, arising from a multitude of factors, leading to scattered interstitial alterations, pulmonary edema, and subsequent acute hypoxic respiratory insufficiency. Acute lung injury (ALI), along with its more serious counterpart, acute respiratory distress syndrome (ARDS), carry a fatality rate that hovers around 30– 40%. Its principal pathological characteristic lies in the unchecked inflammatory reaction. Currently, the main strategies for treating ALI are alleviation of inflammation and prevention of respiratory failure. Concerning the etiology of ALI, NLRP3 Inflammasome is essential to the body’s innate immune response. The composition of this inflammasome complex includes NLRP3, the pyroptosis mediator ASC, and pro-caspase-1. Recent research has reported that the inflammatory response centered on NLRP3 inflammasomes plays a key part in inflammation in ALI, and may hence be a prospective candidate for therapeutic intervention. In the review, we present an overview of the ailment characteristics of acute lung injury along with the constitution and operation of the NLRP3 inflammasome within this framework. We also explore therapeutic strategies targeting the NLRP3 inflammasome to combat acute lung injury.

Keywords: acute lung injury, NLRP3 inflammasome, caspase-1, IL-1β, IL-18

Introduction

The pandemic-causing influenza and emerging viruses have brought about sporadic spikes in global acute lung injury (ALI). ALI manifests through injury to the cells lining the alveoli and those composing the capillary walls, which leads to interstitial and alveolar edema within the lungs, thus leading to a severe shortage of oxygen causing respiratory distress.1,2 ALI presents with reduced pulmonary capacity, diminished elasticity within the lungs, and a profound disruption of the ventilation-perfusion balance.3 When left untreated, acute lung injury (ALI) can develop into acute respiratory distress syndrome (ARDS).4 Despite advancements in grasping the fundamental processes that lead to acute lung injury (ALI), an effective targeted treatment for the condition remains elusive. While ALI morbidity and mortality have declined, the death rate associated with ALI and ARDS continues to hover around a staggering 30 to 40%. It is typified by an unchecked inflammatory reaction. Currently, the main strategies for treating ALI are reduction of inflammation and prevention of respiratory failure. Consequently, gaining a deeper comprehension of the inflammatory origins of ALI and stalling its advancement is crucial.

The NLRP3 inflammasome, recognized as the extensively studied NLR receptor family member, is widely present across various immune cells and holds a crucial part in the body’s immune system, safeguarding the body against the infiltration of infectious agents. This intricate entity comprises a detector (NLRP3), a connector (equipped with the c-terminal cysteine protease enlistment domain, ASC), and an executor (caspase-1). It is a vital element of the innate immune system, enabling the development of active caspase-1 as well as the following development and production of inflammatory cytokines IL-1β and IL-18.5 Important functions in the inflammatory response are played by IL-1β and IL-18.

Activating the NLRP3 inflammasome is a critical stage in the development of ALI. In ALI/ARDS patients, elevated IL-1β and IL-18 levels correlate with unfavorable outcomes. One study found that levels of IL-1β were markedly increased in the BALF of mice with ALI model produced by LPS, and the expression of NLRP3 protein was noticeably higher in lung tissue. The NLRP3 inflammasome acts as an important catalyst and accelerator of ALI. By coordinating the discharge of pro-inflammatory cytokines.6 Consequently, hindering the stimulation of the NLRP3 inflammasome has the potential to be a viable method in managing ALI.

In this review, we offer a succinct overview of ALI, followed by an in-depth look at the make-up and triggering of the NLRP3 inflammasome. We investigate the link between ALI and the NLRP3 inflammasome, as well as potential therapy options for ALI.

Acute Lung Injury

Concept

Since the initial conceptualization of acute respiratory distress syndrome (ARDS) back in 1967, there has been remarkable advancement in unraveling the mechanisms underlying its onset and progression, as well as the physiological changes associated with acute lung injury (ALI).7 ALI presents as a grave medical emergency, carrying a death rate of approximately 30 to 40%. Its characteristics include the aggregation of inflammatory cells in pulmonary tissue, loss of alveolar-capillary membrane integrity, interstitial edema, abnormal gas exchange, and alveolar septal damage. Treatment approaches for ALI remain very limited in their efficacy.8

Diagnostic Criteria

The diagnostic criteria for ALI/ARDS have undergone several changes over the years. In 1988, the system for assessing the degree and severity of lung injury was proposed by Murray et al.9 In 1994, the American-European Consensus Conference (AECC) laid down the definition for ALI/ARDS and set forth the subsequent diagnostic criteria: chest x-rays showing acute bilateral diffuse pulmonary infiltrates; a PaO2/FiO2 ratio ≤300 mmHg for ALI and ≤200 mmHg for ARDS; and either PAWP ≤18 mmHg or absence of indications of left atrial hypertension. This set of criteria first introduced the concept of ALI, with ARDS considered a more severe form of ALI warranting significant attention in the early days of ALI.10 However, the AECC criteria faced many clinical issues. In 2012, a collaborative committee comprising the European Society of Intensive Care Medicine, the American Thoracic Society, and the American Society of Critical Care Medicine unveiled the Berlin Criteria for ARDS. ARDS is categorized into different levels based on the degree of hypoxemia- mild (PaO2/FiO2 is between 200mmHg to 300mmHg), moderate (PaO2/FiO2 is between 100mmHg to 200mmHg), and severe (PaO2/FiO2 is less than or equal to 100mmHg).11 Compared to earlier attempts, the Berlin Criteria provide a better prediction of mortality and do not require measuring pulmonary capillary wedge pressure.

Etiological Factors

The etiology of ALI is complex, with pneumonia being a leading factor causing ARDS and ALI.12 Sepsis and gastric content aspiration are common reasons for pulmonary impairment.13,14 There are also some less common factors, such as massive transfusion, blast waves, smoke inhalation, and toxic gas inhalation.15–18 If two or more of these risk factors are present at the same time, the probability of developing ALI or ARDS becomes higher. For instance, a common cause of sepsis is gram-negative bacterial infection. The outer membrane of gram-negative bacteria harbors lipopolysaccharides (LPS) that can trigger an inflammatory reaction in the body during infections, potentially leading to lung damage.19,20

Pathogenesis

The development of ALI is highly intricate, involving multiple mechanisms such as inflammation activation,6 cell apoptosis,21 oxidative stress injury,22 and coagulation dysfunction.23 It is widely believed that at the core of its pathogenesis is an uncontrolled inflammatory response. The initiation of inflammatory cells and the ensuing discharge of various inflammatory agents in ALI prompted by LPS inflict damage on the alveolar-capillary barrier disruption results in heightened permeability. This pernicious cycle ultimately advances into pulmonary fibrosis, lung injury, and lung edema.24

Signaling Pathways

The inflammatory response during ALI has been shown by recent studies to involve various signaling pathways like AMPK, Wnt/β-catenin, RhoA/ROCK, JAK/STAT, NF-κB, VEGF, PI3K/Akt, and MAPK.(Figure 1).

Figure 1 Signaling pathways implicated in acute lung injury. (A) AMPK - Nrf2 - HO-1; (B) Wnt - β-catenin - TNF-β; (C) RhoA - ROCK – eNOS; (D) JAK - STAT - IL-6, IL-1β, TNF-α; (E) NF-kB - NLRP3 - caspase-1 - IL-1β, IL-18; (F) VEGF - VEGFR2; (G) PI3K - Akt - mTOR - HIF-1α; (H) MAPKs - JNK - NETs; MAPKs - ERK1/2 - NETs; MAPKs - P38.

Abbreviations: AMPK, AMP-activated protein kinase; Nrf2, Nuclear Factor Erythroid 2-Related Factor 2; HO-1, heme oxygenase-1; TNF-β, tumor necrosis factor-β; ROCK, Rho-associated kinase; Enos, Endothelial Nitric Oxide Synthase; JAK, anus kinase; STAT, signal transducer and activator of transcription; IL-6, Interleukin-6; IL-1β, Interleukin-1β; TNF-α, tumor necrosis factor-α; NF-Kb, nuclear factor kappa-B; NLRP3, NOD like receptor heat protein domain related protein 3; IL-18, Interleukin-18; VEGF, Vascular Endothelial Growth Factor; VEGFR2, Vascular Endothelial Growth Factor Receptor 2; PI3K, phosphoinositide 3 kinase; Akt, Protein Kinase B; mTOR, mammalian target of rapamycin; HIF-1α, hypoxia inducible factor-1α; MAPKs, mitogen activated protein kinases; JNK, c-Jun N-terminal kinase; NETs, neutrophil extracellular traps; ERK1/2, extracellular signal-regulated kinase.

AMPK Signaling Pathway

The AMP-activated protein kinase (AMPK) regulates cellular energy metabolism, ensuring energy balance is maintained during metabolic stress.25 The transcription regulator nuclear factor erythroid 2-related factor 2 (Nrf2) is involved in the antioxidant response. In particular, it regulates heme oxygenase-1 (HO-1) expression. Nrf2 acts downstream of AMPK. Activation of Nrf2 and HO-1 by AMPK suppresses reactive oxygen species production.26 Studies have shown that Xanthohumol enhances AMPK-Nrf2 pathway to mitigate LPS-induced ALI in mice.27

Wnt/β-Catenin Signaling Pathway

Wnt family constituents are glycoproteins that exert their action via autocrine or paracrine modes. Wnt interacts with cell surface-specific receptors, triggering a cascade of protein phosphorylation and dephosphorylation events, which result in nuclear β-catenin accumulation.28 Many biological processes such as cellular growth, maturation, programmed cell death, motility, penetration, and tissue system balance depend on the Wnt/β-catenin signaling pathway.29 Villar J. et al30 reported that the Wnt/β-catenin signaling pathway experienced heightened activation in animals suffering from sepsis-triggered ALI. Moreover, defensive MV repressed WNT/β-catenin signaling pathway and enhanced pulmonary restoration.

RhoA/ROCK Signaling Pathway

The RhoA, a diminutive GTPase protein belonging to the Rho family, represents a key component known as the Ras homologous gene family member A. The Rho-Associated Protein Kinase (ROCK) functions as a downstream effector of RhoA. The Rho/ROCK signaling pathway is involved in cell growth, maturation, movement, and evolution.31 Endothelial NO synthase (eNOS) can mediate LPS-induced RhoA activation, resulting in pulmonary dysfunction and pro-inflammatory cytokine secretion. Blocking eNOS activity can suppress NF-κB signal transduction.32 Research has demonstrated the engagement of the Rho/ROCK signaling cascade serves as a mediator for increased permeability in pulmonary endothelial tissues, and it holds a vital role in the pathophysiological progression of ALI.33

JAK/STAT Signaling Pathway

Janus kinase (JAK) is an intracellular tyrosine kinase that plays a crucial role in signal transduction initiated via multiple membrane receptors. The JAK/STAT signaling cascade governs a myriad of cell functions that are vital for maintaining internal equilibrium.34 It also contributes to the emergence of a wide array of inflammatory and autoimmune disorders.35 Zhang et al36 showed that during sepsis, LPS upregulates JAK/STAT expression, interacting with Toll-like receptors to cause the excretion of IL-10, IL-6, and TNF-α, resulting in alveolar epithelial and vascular endothelial cell injury, leading to diffuse ALI.

NF-kB Signaling Pathway

NF-κB acts as a transcriptional activator, spurring the creation of cytokines and enhancing cellular longevity. NF-κB exists in nearly every type of animal cell, engaging in cellular reactions to stimuli, such as cytokines, stress, oxidative LDL, free radicals, and viral antigens or bacterial.37 Several investigations have demonstrated that ALI is linked to the start and activation of NLRP3 inflammasomes, which damage and inflame the lungs via the NF-kB pathway. The subsequent triggering of caspase-1 leads to the release and development of IL-1β and IL-18, which drive cell apoptosis and inflammatory reactions, thus aggravating ALI.38–44

VEGF Signaling Pathway

The angiogenic stimulant vascular endothelial growth factor (VEGF) encourages the expansion of endothelial cells. It stimulates the proliferation, increases the permeability, and enhances endothelial cell migration.45 Research has indicated that pulmonary VEGF upregulation plays a crucial part in lung damage caused by fat embolism. VEGF expression is increased in several forms of ARDS, and its severity is associated with mortality. VEGFR-2 antagonists can significantly alleviate lung inflammation and cellular damage induced by fat embolisms.46

PI3K/Akt Signaling Pathway

The PI3K/AKT cascade functions as an internal messenger system that reacts to external cues, enhancing cellular metabolism, division, survival, development, and the formation of new blood vessels.47 In addition, the PI3K/Akt signaling pathway performs a vital function in cellular survival and resistance to oxidative stress during pulmonary inflammation.48 Li et al49 revealed that within mature dendritic cells, HMGB1 triggers the PI3K/Akt/mTOR signaling cascade, upregulating the mRNA levels and subsequent activation of PI3K, Akt, and mTOR to facilitate the development of lung inflammation in ALI.

MAPK Signaling Pathway

Mitogen activated protein kinase (MAPK) comprises p38, ERK and JNK subfamilies, which are key regulatory factors in cell physiology and the pathogenesis of various diseases, including cancer, where they regulate cell proliferation, growth, and apoptosis.50 Neutrophil extracellular traps (NETs) indirectly induced by LPS cause acute lung injury.51 Erythropoietin may provide relief from acute lung injury resulting from ischemia-reperfusion by inhibiting the p38 MAPK signaling pathway.52 FK866 can partially inhibit the JNK pathway and decreases reactive oxygen species production in neutrophils, thereby suppressing NETs.53 Redouning improves ALI by obstructing the phosphorylation process of ERK1/2 and the formation of NETs.54

NLRP3 Inflammasome

Overview

Innate immunity acts as the initial defense mechanism against the incursion of pathogens. Unlike the adaptable branch of the immune system, it has nonspecific defense functions.55 Innate immunity comprises two lines. The first one includes natural barriers effectively prevent the invasion of common pathogens into tissue.56 The second line consists of leukocytes, which eliminate pathogens upon detection.57 Pattern recognition receptors (PRRs) expressed in germ cells are primarily responsible for the primary stimulation of inherent immune responses. PRRs can detect the presence of microbes by recognizing conserved pathogen macromolecular structures, known as pathogen-associated molecular patterns (PAMPs). They are also capable of recognizing endogenous substances secreted by compromised cells, commonly known as damage-associated molecular patterns (DAMPs).58 In response to infection, PRRs activate signaling pathways, thereby stimulating the host defense response against microbial invasion.59 PRRs can be classified into numerous categories, which encompass TLRs, RIG-I-like receptors, and nucleotide-binding oligomerization domain receptors and leucine-rich repeat-containing receptors (NLRs), synthase of cyclic GMP-AMP and the route that activates interferon genes, along with AIM2-similar receptors and C-type lectin.60 When these PRRs are activated, they can initiate an inflammatory reaction to combat infections and heal tissue injuries. Through these mechanisms, innate immunity plays ensures an effective response to harmful external stimuli.

Intracellular pattern recognition receptors (PRRs) orchestrate the assembly of inflammasomes, which are intricate multi-protein structures.61 Once assembled, inflammasomes can trigger the enzymatic activity of caspase-1 advances the development and release of IL-1β and IL-18, in addition to cleaving gasdermin-D (GSDMD) and thus facilitating cellular pore formation and subsequent pyroptosis.62 The NLRP3 is the most thoroughly examined inflammasome.63 It is primarily expressed in macrophages and neutrophils. Abnormal NLRP3 inflammasome activation can cause inflammatory ailments.64

Composition of the NLRP3 Inflammasome

The effector enzyme caspase-1, adaptor ASC (sometimes referred to as PYCARD), and sensor NLRP3 make up the NLRP3 inflammasome.65 The NLRP3 molecule is composed of three unique segments: an N-terminal pyrin domain, a NACHT domain situated centrally, and a C-terminal domain that is characterized by a series of leucine-rich repeats. The NACHT domain plays a pivotal role in driving ATPase activity, an essential mechanism for the self-assembly and operational efficacy of NLRP3.66 This domain is also a major structural component of the inflammasome. ASC is comprised of two integral components: the PYD domain, which associates with NLRP3, and the CARD domain, which facilitates its connection to caspase-1.67 The effector caspase-1 of the NLRP3 inflammasome converts IL-1β and IL-18 precursors into their activated forms.68 Caspase-1 plays a pivotal role in cell death associated with inflammation, specifically through a process known as pyroptosis.69

NLRP3 Inflammasome activation

The initiation of NLRP3 inflammasome entails a dual approach: firstly, a preparatory phase, and secondly, the actual triggering event.70 The NLRP3 inflammasome activation entails four typical pathways: ion channels, mitochondrial autophagy, excessive reactive oxygen species (ROS) production and lysosomal rupture.71

Priming of the NLRP3 Inflammasome

During the priming phase, immune cells must be exposed to inducing stimuli such as LPS and tumor necrosis factors, which bind to transmembrane PRRs such as IL-1R, TLRs, and TNFR. This binding activates NF-kB, further upregulating the expression of inflammasome components NLRP3 and pro-IL-1β. The presence of high levels of NLRP3 and pro-IL-1β is crucial for inflammasome formation.72

Activation of the NLRP3 Inflammasome

Upon the first signal, NLRP3 is stimulated by diverse activators including inherent DAMPs, assorted PAMPs, multiple pathogens, and potassium ions.73 The structural proteins of NLRP3 bind to the PYD of the adaptor ASC through oligomerization. Next, the CARD domain within ASC interacts with the CARD in pro-caspase-1, culminating in the formation of the fully integrated NLRP3 inflammasome, and thereby converting pro-caspase-1 to its active form caspase-1. IL-1β and IL-18 mature as a result of caspase-1 activation, and GSDMD is cleaved into its N- and C-terminal components. The GSDMD N-terminus can form pores by binding to the cell membrane, thus promoting the secretion of pro-inflammatory cytokines (IL-1β and IL-18), which in turn, initiates pyroptotic cell death. (Figure 2).74,75

Figure 2 Mechanisms of NLRP3 inflammasome activation. The activation of NLRP3 inflammasome occurs via four classical pathways: (1) ion channels; (2) excessive production of reactive oxygen species; (3) mitochondrial autophagy, and (4) lysosomal rupture. Activation of the NLRP3 inflammasome occurs in two phases. In the priming phase, a stimulus binds to pattern recognition receptors and activates NF-kB to further upregulate the expression of NLRP3, pro-IL-1β, and pro-IL-18. In the activation phase, the structural protein NLRP3 binds to the PYD of ASC through oligomerization, whereafter the CARD of ASC binds to the CARC of pro-caspase-1 to form the intact NLRP3 inflammasome. The NLRP3 inflammasome then activates pro-caspase-1, which in turn promotes the maturation of IL-1β and IL-18, in addition to cleaving Gasdermin-D to N- and C-terminal fragments, with the activated N terminus promoting pyroptosis.

NLRP3 Inflammasome and ALI

There is considerable proof highlighting the crucial function of the NLRP3 inflammasome in ALI.38 It has been demonstrated through research that patients suffering from ARDS often exhibit increased concentrations of IL-1β and IL-18, markers that typically correlate with unfavorable outcomes.39 Injection of extracellular mitochondrial DNA into the trachea of mice, NLRP3 inflammasome is promoted through the TLR9, p38MAPK, and NF-κB pathways, which leads to increased expression of NLRP3, ASC, and caspase-1. This in turn stimulates IL-1β and IL-18 release to cause lung injury.40 In a murine model of LPS-induced septic ALI, IL-1β and IL-18 concentrations in bronchoalveolar lavage fluid (BALF) and lung tissue were markedly elevated, coinciding with an uptick in the activity of NLRP3 and caspase-1.41 The upregulation of inflammatory cytokines IL-18, IL-1β, and TNF-α is observed in ALI induced by sepsis, indicating that Activation of NLRP3 inflammasome stimulates generation of proinflammatory cytokines, and its manifestation is correlated positively with disease severity and fatality.42 In ventilator-induced lung damage, TLR4 activation activates NLRP3 inflammasome, thereby promoting inflammatory damage. Further, NLRP3 knockout significantly alleviates ventilator-induced lung injury.43,44 In summary, Numerous investigations have indicated a close link between ALI and the NLRP3 inflammasome. Targeted suppression of NLRP3 inflammasome and related signaling pathways represents a new direction in ALI prevention and treatment research.

Therapeutic Approaches

Research on ways to produce anti-inflammatory medications that hinder the NLRP3 inflammasome activation has become a focal point of interest in recent research. Given the link between ALI and the NLRP3 inflammasome, targeting NLRP3 for inhibition has increasingly become a pivotal approach in the therapeutic intervention for ALI.

Inhibiting the NF-kB Pathway

Propofol is a commonly used drug for anesthesia induction and maintenance. Propofol is also known to harbor anti-cancer,76 antioxidant,77 neuroprotective,78 and anti-inflammatory activities.79 Prior research has demonstrated that propofol influences pulmonary damage. Exposure to sevoflurane and propofol exhibits anti-inflammatory properties in the context of LPS-triggered ALI.80 Propofol has been shown to reduce inflammation and oxidative stress by inhibiting the p38 MAPK/NF-κB pathway and NLRP3 inflammasome activation, as well as to alleviate LPS-induced pulmonary edema in neonatal rats. Propofol effectively reversed the effects of LPS treatment on the NLRP3 inflammasome and the p38 MAPK/NF-κB pathway in newborn rats.81 Propofol may thus be a prospective medication for the therapy of newborn ALI.

Metformin is an established and effective anti-diabetic drug. It additionally demonstrates protective benefits for the lungs across various acute lung injury scenarios,82 PM2.5-induced83 and endotoxemia-induced pulmonary injury.84 Its action may include the activation of AMP-activated protein kinase (AMPK) to suppress NF-κB in endothelial cells and consequently inhibit cytokine-induced inflammation and adhesion factor expression.85 That is, metformin mitigates LPS-triggered ALI by inhibiting endothelial cell pyroptosis by suppressing the NF-κB-NLRP3 signaling pathway. Throughout the progression of inflammatory lung injury induced by LPS, the pulmonary endothelium undergoes impairment, rendering it unable to effectively maintain a protective barrier. Metformin effectively counteracts the negative repercussions by reversing them, thus safeguarding the endothelial pathways and ameliorating lung function.86 Glibenclamide also has the ability to reduce LPS-induced ALI damage.87

As an anti-inflammatory agent, berberine has therapeutic effects in many diseases. Berberine modulates the NF-κB signaling pathway, exhibiting anti-inflammatory properties.88 It can suppress the triggering of the NLRP3 inflammasome by the influenza virus in macrophages by enhancing mitochondrial autophagy and reducing mitochondrial ROS, thus mitigating lung damage.89 Moreover, berberine has the ability to inhibit the interaction of NLRP3 with NEK7, resulting in a direct and effective anti-inflammatory response.90 Berberine treatment down-regulated p-NF-κB, further suppressing NLRP3 levels in vivo and in vitro. This caused a marked reduction in IL-18 and IL-1β concentrations in pulmonary tissues, considerably alleviating inflammation.91 These results underscore the promising role that berberine could play in treating ALI.

Glycyrrhizic acid is renowned for its excellent pharmacological effects and various biological activities, including antiviral and anti-inflammatory properties.92 Glycyrrhizic acid can reduce the production of IL-Iβ and TNF-α in inflammation, inhibiting NF-κB activation.93 Furthermore, it may thwart colorectal cancer development by disrupting the HMGB1-TLR4-NF-κB signaling cascade.94 Glycyrrhizic acid modulates autophagy through the PI3K/AKT/mTOR pathway to improve LPS-induced ALI.95 It possesses the capability to modulate the signaling cascade of the NF-κB/NLRP3 inflammasome, hence curtailing the inflammatory reaction instigated by LPS.96 To sum it up, glycyrrhizic acid may alleviate ALI by suppressing the NF-kB pathway, positioning it as a potential therapeutic agent.

Remarkably, it has also been demonstrated that LPS-induced acute lung injury can be relieved by isochlorogenic acid A,97 pterin,98 and hederasaponin-C99 through modification of the NF-κB-NLRP3 signal pathways.

Inhibiting Excessive Production of ROS

Oxidative stress, coupled with the ensuing inflammation, are key underlying pathological mechanisms in ALI. Emodin, an active constituent of Rhubarb, has anti-inflammatory characteristics. Studies have shown that emodin can improve LPS-induced ALI.100,101 Furthermore, emodin has the ability to suppress ROS, MPO, and MDA generation. In addition, emodin guards against ALI brought on by LPS, presumably by lowering ROS generation and suppressing NLRP3 expression.102 Emodin therefore represents a prospective medical remedy for ALI and lung inflammation.

Nrf2 occupies a pivotal role as a transcription factor in the antioxidant response, and it comes into action by inhibiting oxidative stress and inflammation.103 Recent research indicates that Nrf2 can prevent activation of the NLRP3 inflammasome.104 Numerous substances possess the ability to alleviate LPS-triggered ALI through Nrf2 downregulation of NLRP3 inflammasome-induced cellular apoptosis. Among them, honokiol, a compound extracted from the Magnolia officinalis tree, is known for its abilities to combat oxidative stress and inflammation. HKL markedly increases the protein and mRNA levels of Nrf2 and HO-1, reduces MPO and MDA levels, and boosts SOD levels in vivo and in vitro. Moreover, Nrf2 knockdown can reverse the antioxidant effect of HKL, indicating that the latter exerts its antioxidant effect in LPS-induced ALI via Nrf2/HO-1 signaling.105 Oridonin (Ori), a natural substance with antioxidant and anti-inflammatory effects extracted from Rabdosia rubescens, serves as an activator of Nrf2. It can reduce LPS-triggered inflammation through the NF-κB pathway.106 In addition, it generates a covalent connection with cysteine 279 situated in the NACHT domain of NLRP3, thus impeding the interaction and activation of the NLRP3 inflammasome by hindering the link between NLRP3 and NEK7.107 Overall, Ori delivers protective benefits against LPS induced acute lung injury by activating its anti-inflammatory and antioxidant responses through the Nrf2 pathway. Moreover, compounds such as melatonin,108 hydnocarpin D,109 isoorientin,110 citrulline,111 and glycyrrhizic acid95 also inhibit NLRP3 inflammasome-mediated cell apoptosis through Nrf2, reducing LPS-induced ALI.

Inhibiting Mitochondrial Autophagy

Mitochondrial autophagy is notably related to NLRP3 inflammasome activation.112 Sestrin2 (Sesn2), a highly conserved stress-induced protein, plays a vital part in regulating cellular stress reactions and antioxidant protection.113 Studies have shown that Sesn2 can prevent sepsis by inducing mitochondrial autophagy and suppressing NLRP3 activation in macrophages.114 Moreover, Sesn2 has the ability to inhibit the NLRP3 inflammasome’s activation, which is typically instigated by LPS within pulmonary macrophages, reducing cell apoptosis and protecting mitochondria from damage. Sesn2 maintains mitochondrial homeostasis in macrophages through the Pink1/Parkin signaling pathway during mitochondrial autophagy, ultimately protecting the lungs from LPS-induced ALI.115 These discoveries validate Sesn2 as a fresh therapeutic focus for managing ALI/ARDS.

Inhibiting Ion Channels

Calcium, a crucial intracellular secondary messenger, plays a role in several cellular mechanisms.116 The influx of calcium ions is recognized to contribute to NLRP3 inflammasome activation.117 The binding of calcium to calmodulin (CaM) leads to activation of CaM kinase (CaMK), which initiates inflammation.118 In alveolar type II epithelial cells, CaMK4 is essential for initiating the NLRP3 inflammasome, which exacerbates lung damage in mice with LPS-induced ALI. The drug KN-93, a CaMK4 inhibitor, can effectively improve ALI by inhibiting NLRP3 inflammasome activation.119 Thus, inhibiting CaMK4 could represent a new approach for treating ALI.

Inhibiting the Maturation and Secretion of IL-1β and IL-18

The NLRP3 inflammasome coordinates IL-1β and IL-18 processing and secretion. Blocking IL-1β and IL-18 production or upstream signaling may be effective in treating ALI/ARDS.120 Rapamycin, a natural product with immunosuppressive effects widely used in patients who undergo organ transplantation,121 has been confirmed to suppress autophagy by inhibiting mTOR an thus regulating the production of IL-1β and IL-18.122 The induction of autophagy can limit the secretion of IL-1β and IL-18 by clearing damaged mitochondria and preventing the release of mitochondrial ROS.123,124 Therefore, rapamycin-induced autophagy helps to reduce the generation of IL-1β and IL-18 after LPS exposure. It has also been demonstrated that rapamycin treatment reduces the total number of cells as well as the neutrophil count in BALF after an LPS infection.125,126 Further, rapamycin shields mice from lung damage triggered by LPS by suppressing mTOR activity, which subsequently reduces IL-1β and IL-18 production, suppressing immune cell infiltration.127 Therefore, rapamycin may represent an effective drug for treating ALI (Table 1).

Table 1 Summary of Drugs That Inhibit the NLRP3 Inflammasome for the Treatment of Acute Lung Injury

Summary

Herein, we reviewed the progress in our understanding of ALI and the configuration and stimulation of NLRP3 inflammasome, and explored the NLRP3 inflammasome’s mechanism and potential therapeutic approaches in ALI. The NLRP3 inflammasome has the potential to be an ideal candidate for early detection and treatment of ALI owing to its crucial functions in transducing signals and releasing pro-inflammatory cytokines. Future investigations ought to prioritize the identification of upstream factors that affect the NLRP3 inflammasome and the development of interventions aimed at its assembly. Taken together, targeting NLRP3-driven inflammation may represent an important direction to treat and prevent ALI.

Acknowledgments

This work was supported by Liaoning Provincial Applied Basic Research Program Joint Project (2022JH2/101500074). The figures were supported by Figdraw.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Mowery NT, Terzian WTH, Nelson AC. Acute lung injury. Curr Prob Surg. 2020;57(5):100777. doi:10.1016/j.cpsurg.2020.100777

2. Lu Q, Yu S, Meng X, et al. MicroRNAs: important regulatory molecules in acute lung injury/acute respiratory distress syndrome. IJMS. 2022;23(10):5545. doi:10.3390/ijms23105545

3. Mokra D. Acute lung injury – from pathophysiology to treatment. Physiol Res. 2020;69(Suppl 3):S353–S366. doi:10.33549/physiolres.934602

4. Liu C, Xiao K, Xie L. Advances in the use of exosomes for the treatment of ALI/ARDS. Front Immunol. 2022;13:971189. doi:10.3389/fimmu.2022.971189

5. Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. IJMS. 2019;20(13):3328. doi:10.3390/ijms20133328

6. Fukatsu M, Ohkawara H, Wang X, et al. The suppressive effects of Mer inhibition on inflammatory responses in the pathogenesis of LPS-induced ALI/ARDS. Sci Signal. 2022;15(724):eabd2533. doi:10.1126/scisignal.abd2533

7. Matthay MA, Zimmerman GA. Acute lung injury and the acute respiratory distress syndrome: four decades of inquiry into pathogenesis and rational management. Am J Respir Cell Mol Biol. 2005;33(4):319–327. doi:10.1165/rcmb.F305

8. Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J Clin Invest. 2012;122(8):2731–2740. doi:10.1172/JCI60331

9. Murray JF, Matthay MA, Luce JM, Flick MR. An expanded definition of the adult respiratory distress syndrome. Am Rev Respir Dis. 1988;138(3):720–723. doi:10.1164/ajrccm/138.3.720

10. Bernard GR, Artigas A, Brigham KL, et al.; The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med. 1994;149(3):818–824. doi:10.1164/ajrccm.149.3.7509706

11. Ranieri VM, Rubenfeld GD, Taylor Thompson B, et al. Acute respiratory distress syndrome: the berlin definition. JAMA. 2012;307(23). doi:10.1001/jama.2012.5669

12. Long ME, Mallampalli RK, Horowitz JC. Pathogenesis of pneumonia and acute lung injury. Clin Sci. 2022;136(10):747–769. doi:10.1042/CS20210879

13. Kumar V. Pulmonary innate immune response determines the outcome of inflammation during pneumonia and sepsis-associated acute lung injury. Front Immunol. 2020;11:1722. doi:10.3389/fimmu.2020.01722

14. Ayala P, Vivar R, Montalva R, Olmos P, Meneses M, Borzone GR. Elastin degradation products in acute lung injury induced by gastric contents aspiration. Respir Res. 2018;19(1):165. doi:10.1186/s12931-018-0873-1

15. Mulder HD, Augustijn QJJ, Van Woensel JB, Bos AP, Juffermans NP, Wösten-van Asperen RM. Incidence, risk factors, and outcome of transfusion-related acute lung injury in critically ill children: a retrospective study. J Crit Care. 2015;30(1):55–59. doi:10.1016/j.jcrc.2014.10.005

16. Mathews ZR, Koyfman A. Blast Injuries. J Emerg Med. 2015;49(4):573–587. doi:10.1016/j.jemermed.2015.03.013

17. Zhang F, Li M, Lan Y, Wang C. Imbalance of Th17/Tregs in rats with smoke inhalation-induced acute lung injury. Sci Rep. 2016;6(1):21348. doi:10.1038/srep21348

18. Cao C, Zhang L, Shen J. Phosgene-Induced acute lung injury: approaches for mechanism-based treatment strategies. Front Immunol. 2022;13:917395. doi:10.3389/fimmu.2022.917395

19. Hsieh YH, Deng JS, Pan HP, Liao JC, Huang SS, Huang GJ. Sclareol ameliorate lipopolysaccharide-induced acute lung injury through inhibition of MAPK and induction of HO-1 signaling. Int Immunopharmacol. 2017;44:16–25. doi:10.1016/j.intimp.2016.12.026

20. Kolomaznik M, Nova Z, Calkovska A. Pulmonary surfactant and bacterial lipopolysaccharide: the interaction and its functional consequences. Physiol Res. 2017;S147–S157. doi:10.33549/physiolres.933672

21. Liu C, Zhou Y, Tu Q, Yao L, Li J, Yang Z. Alpha-linolenic acid pretreatment alleviates NETs-induced alveolar macrophage pyroptosis by inhibiting pyrin inflammasome activation in a mouse model of sepsis-induced ALI/ARDS. Front Immunol. 2023;14:1146612. doi:10.3389/fimmu.2023.1146612

22. Puri G, Naura AS. Critical role of mitochondrial oxidative stress in acid aspiration induced ALI in mice. Toxicol Mech Methods. 2020;30(4):266–274. doi:10.1080/15376516.2019.1710888

23. Glas GJ, Van Der S, Schultz MJ, Hofstra J‐J, Van Der Poll T, Levi M. Bronchoalveolar hemostasis in lung injury and acute respiratory distress syndrome. J Thromb Haemost. 2013;11(1):17–25. doi:10.1111/jth.12047

24. Jiang Q, Yi M, Guo Q, et al. Protective effects of polydatin on lipopolysaccharide-induced acute lung injury through TLR4-MyD88-NF-κB pathway. Int Immunopharmacol. 2015;29(2):370–376. doi:10.1016/j.intimp.2015.10.027

25. Kim J, Yang G, Kim Y, Kim J, Ha J. AMPK activators: mechanisms of action and physiological activities. Exp Mol Med. 2016;48(4):e224–e224. doi:10.1038/emm.2016.16

26. Shen B, Zhao C, Wang Y, et al. Aucubin inhibited lipid accumulation and oxidative stress via Nrf2/ HO ‐1 and AMPK signalling pathways. J Cell Mole Med. 2019;23(6):4063–4075. doi:10.1111/jcmm.14293

27. Lv H, Liu Q, Wen Z, Feng H, Deng X, Ci X. Xanthohumol ameliorates lipopolysaccharide (LPS)-induced acute lung injury via induction of AMPK/GSK3β-Nrf2 signal axis. Redox Biol. 2017;12:311–324. doi:10.1016/j.redox.2017.03.001

28. Liu J, Xiao Q, Xiao J, et al. Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Sig Transduct Target Ther. 2022;7(1):3. doi:10.1038/s41392-021-00762-6

29. Zhang Y, Wang X. Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol. 2020;13(1):165. doi:10.1186/s13045-020-00990-3

30. Villar J, Cabrera NE, Casula M, et al. WNT/β-catenin signaling is modulated by mechanical ventilation in an experimental model of acute lung injury. Intensive Care Med. 2011;37(7):1201–1209. doi:10.1007/s00134-011-2234-0

31. Deng Z, Jia Y, Liu H, et al. RhoA/ROCK pathway: implication in osteoarthritis and therapeutic targets. Am J Transl Res. 2019;11(9):5324–5331.

32. Wang H, Sun X, Lu Q, et al. The mitochondrial redistribution of eNOS is involved in lipopolysaccharide induced inflammasome activation during acute lung injury. Redox Biol. 2021;41:101878. doi:10.1016/j.redox.2021.101878

33. Han J, Ding R, Zhao D, Zhang Z, Ma X. Unfractionated heparin attenuates lung vascular leak in a mouse model of sepsis: Role of RhoA/Rho kinase pathway. Thromb Res. 2013;132(1):e42–e47. doi:10.1016/j.thromres.2013.03.010

34. Montero P, Milara J, Roger I, Cortijo J. Role of JAK/STAT in interstitial lung diseases; molecular and cellular mechanisms. IJMS. 2021;22(12):6211. doi:10.3390/ijms22126211

35. Banerjee S. JAK–STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs. 2017;77:521–546.

36. Zhang X, Wang X, Sun L, Gao G, Li Y. Tofacitinib reduces acute lung injury and improves survival in a rat model of sepsis by inhibiting the JAK-STAT/NF-κB pathway. J Inflamm. 2023;20(1):5. doi:10.1186/s12950-023-00332-3

37. De Luca F. Regulatory role of NF-κB in growth plate chondrogenesis and its functional interaction with Growth Hormone. Molec Cell Endocrinol. 2020;514:110916. doi:10.1016/j.mce.2020.110916

38. Lei J, Shen Y, Xv G, Di Z, Li Y, Li G. Aloin suppresses lipopolysaccharide-induced acute lung injury by inhibiting NLRP3/NF-κB via activation of SIRT1 in mice. Immuno and Immunotoxicology. 2020;42(4):306–313. doi:10.1080/08923973.2020.1765373

39. Dolinay T, Kim YS, Howrylak J, et al. Inflammasome-regulated cytokines are critical mediators of acute lung injury. Am J Respir Crit Care Med. 2012;185(11):1225–1234. doi:10.1164/rccm.201201-0003OC

40. Wu G, Zhu Q, Zeng J, et al. Extracellular mitochondrial DNA promote NLRP3 inflammasome activation and induce acute lung injury through TLR9 and NF-κB. J Thorac Dis. 2019;11(11):4816–4828. doi:10.21037/jtd.2019.10.26

41. Yang J, Yang J, Huang X, et al. Glibenclamide alleviates LPS-induced acute lung injury through NLRP3 inflammasome signaling pathway. Mediators Inflammation. 2022;2022:1–12. doi:10.1155/2022/8457010

42. Zhang XP, Zhang WT, Qiu Y, et al. Cyclic helix B peptide alleviates sepsis-induced acute lung injury by downregulating NLRP3 inflammasome activation in alveolar macrophages. Int Immunopharmacol. 2020;88:106849. doi:10.1016/j.intimp.2020.106849

43. Kuipers MT, Aslami H, Janczy JR, et al. Ventilator-induced lung injury is mediated by the NLRP3 Inflammasome. Anesthesiology. 2012;116(5):1104–1115. doi:10.1097/ALN.0b013e3182518bc0

44. Dai H, Pan L, Lin F, Ge W, Li W, He S. Mechanical ventilation modulates Toll-like receptors 2, 4, and 9 on alveolar macrophages in a ventilator-induced lung injury model. J Thoracic Dis. 2015;7(4):616–624. doi:10.3978/j.issn.2072-1439.2015.02.10

45. Melincovici CS, Boşca AB, Şuşman S, et al. Vascular endothelial growth factor (VEGF) – key factor in normal and pathological angiogenesis. Rom J Morphol Embryol. 2018;59(2):455–467.

46. Lin CK, Lin YH, Huang TC, Shi CS, Yang CT, Yang YL. VEGF mediates fat embolism-induced acute lung injury via VEGF receptor 2 and the MAPK cascade. Sci Rep. 2019;9(1):11713. doi:10.1038/s41598-019-47276-4

47. Long HZ, Cheng Y, Zhou ZW, Luo HY, Wen DD, Gao LC. PI3K/AKT signal pathway: a target of natural products in the prevention and treatment of alzheimer’s disease and parkinson’s disease. Front Pharmacol. 2021;12:648636. doi:10.3389/fphar.2021.648636

48. Meng L, Li L, Lu S, et al. The protective effect of dexmedetomidine on LPS-induced acute lung injury through the HMGB1-mediated TLR4/NF-κB and PI3K/Akt/mTOR pathways. Mol Immunol. 2018;94:7–17. doi:10.1016/j.molimm.2017.12.008

49. Li R, Zou X, Huang H, et al. HMGB1/PI3K/Akt/mTOR signaling participates in the pathological process of acute lung injury by regulating the maturation and function of dendritic cells. Front Immunol. 2020;11:1104. doi:10.3389/fimmu.2020.01104

50. Lu M, Wang Y, Zhan X. The MAPK pathway-based drug therapeutic targets in pituitary adenomas. Front Endocrinol. 2019;10:330. doi:10.3389/fendo.2019.00330

51. Liu S, Su X, Pan P, et al. Neutrophil extracellular traps are indirectly triggered by lipopolysaccharide and contribute to acute lung injury. Sci Rep. 2016;6(1):37252. doi:10.1038/srep37252

52. Jia L, Cui W, Chen J, et al. Erythropoietin alleviates acute lung injury induced by ischemia-reperfusion through blocking p38 MAPK signaling. Hum Exp Toxicol. 2021;40(12_suppl):S593–S602. doi:10.1177/09603271211043480

53. Ding J, Zhang Z, Huang W, Bi G. Nicotinamide phosphoribosyltransferase inhibitor is a novel therapeutic candidate in LPS‐induced neutrophil extracellular traps. Microbiol Immunol. 2021;65(7):257–264. doi:10.1111/1348-0421.12885

54. Yang C, Song C, Liu Y, et al. Re-Du-Ning injection ameliorates LPS-induced lung injury through inhibiting neutrophil extracellular traps formation. Phytomedicine. 2021;90:153635. doi:10.1016/j.phymed.2021.153635

55. Place DE, Kanneganti TD. The innate immune system and cell death in autoinflammatory and autoimmune disease. Curr Opin Immunol. 2020;67:95–105. doi:10.1016/j.coi.2020.10.013

56. Riera Romo M, Pérez-Martínez D, Castillo Ferrer C. Innate immunity in vertebrates: an overview. Immunology. 2016;148(2):125–139. doi:10.1111/imm.12597

57. Lacy P, Stow JL. Cytokine release from innate immune cells: association with diverse membrane trafficking pathways. Blood. 2011;118(1):9–18. doi:10.1182/blood-2010-08-265892

58. Li D, Wu M. Pattern recognition receptors in health and diseases. Sig Transduct Target Ther. 2021;6(1):291. doi:10.1038/s41392-021-00687-0

59. Hayward JA, Mathur A, Ngo C, Man SM. Cytosolic recognition of microbes and pathogens: inflammasomes in action. Microbiol Mol Biol Rev. 2018;82(4):e00015–18. doi:10.1128/MMBR.00015-18

60. Wicherska-Pawłowska K, Wróbel T, Rybka J. Toll-Like Receptors (TLRs), NOD-Like Receptors (NLRs), and RIG-I-Like Receptors (RLRs) in Innate Immunity. TLRs, NLRs, and RLRs Ligands as Immunotherapeutic Agents for Hematopoietic Diseases. IJMS. 2021;22(24):13397. doi:10.3390/ijms222413397

61. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–832. doi:10.1016/j.cell.2010.01.040

62. Zhang P, Liu Y, Hu L, et al. NLRC4 inflammasome–dependent cell death occurs by a complementary series of three death pathways and determines lethality in mice. Sci Adv. 2021;7(43):eabi9471. doi:10.1126/sciadv.abi9471

63. Huot-Marchand S, Nascimento M, Culerier E, et al. Cigarette smoke-induced gasdermin D activation in bronchoalveolar macrophages and bronchial epithelial cells dependently on NLRP3. Front Immunol. 2022;13:918507. doi:10.3389/fimmu.2022.918507

64. Takahashi M. NLRP3 inflammasome as a key driver of vascular disease. Cardiovasc Res. 2022;118(2):372–385. doi:10.1093/cvr/cvab010

65. Swanson KV, Deng M, Ting JPY. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–489. doi:10.1038/s41577-019-0165-0

66. Freeman TL, Swartz TH. Targeting the NLRP3 Inflammasome in Severe COVID-19. Front Immunol. 2020;11:1518. doi:10.3389/fimmu.2020.01518

67. Lu A, Magupalli VG, Ruan J, et al. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 2014;156(6):1193–1206. doi:10.1016/j.cell.2014.02.008

68. Sun Q, Scott MJ. Caspase-1 as a multifunctional inflammatory mediator: noncytokine maturation roles. J Leukocyte Biol. 2016;100(5):961–967. doi:10.1189/jlb.3MR0516-224R

69. Miao EA, Rajan JV, Aderem A. Caspase‐1‐induced pyroptotic cell death. Immunol Rev. 2011;243(1):206–214. doi:10.1111/j.1600-065X.2011.01044.x

70. Li Z, Guo J, Bi L. Role of the NLRP3 inflammasome in autoimmune diseases. Biomed Pharmacother. 2020;130:110542. doi:10.1016/j.biopha.2020.110542

71. Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128. doi:10.1038/s41419-019-1413-8

72. Song N, Li T. Regulation of NLRP3 Inflammasome by Phosphorylation. Front Immunol. 2018;9:2305. doi:10.3389/fimmu.2018.02305

73. Seok JK, Kang HC, Cho YY, Lee HS, Lee JY. Regulation of the NLRP3 inflammasome by post-translational modifications and small molecules. Front Immunol. 2021;11:618231. doi:10.3389/fimmu.2020.618231

74. Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol. 2021;22(5):550–559. doi:10.1038/s41590-021-00886-5

75. Zhang Y, Yang W, Li W, Zhao Y. NLRP3 inflammasome: checkpoint connecting innate and adaptive immunity in autoimmune diseases. Front Immunol. 2021;12:732933. doi:10.3389/fimmu.2021.732933

76. Wu KC, Liao KS, Yeh LR, Wang YK. Drug Repurposing: The Mechanisms and Signaling Pathways of Anti-Cancer Effects of Anesthetics. Biomedicines. 2022;10(7):1589. doi:10.3390/biomedicines10071589

77. Han J, Tao W, Cui W, Chen J. Propofol via Antioxidant Property Attenuated Hypoxia-Mediated Mitochondrial Dynamic Imbalance and Malfunction in Primary Rat Hippocampal Neurons. Oxid Med Cell Longev. 2022;2022:6298786. doi:10.1155/2022/6298786

78. Kotani Y, Shimazawa M, Yoshimura S, Iwama T, Hara H. The experimental and clinical pharmacology of propofol, an anesthetic agent with neuroprotective properties. CNS Neurosci Ther. 2008;14(2):95–106. doi:10.1111/j.1527-3458.2008.00043.x

79. Vasileiou I, Xanthos T, Koudouna E, et al. Propofol: a review of its non-anaesthetic effects. Eur J Pharmacol. 2009;605:(1–3):1–8. doi:10.1016/j.ejphar.2009.01.007

80. Liu W, Zhu H, Fang H. Propofol potentiates sevoflurane-induced inhibition of nuclear factor--κB-Mediated inflammatory responses and regulation of mitogen-activated protein kinases pathways via toll-like receptor 4 signaling in lipopolysaccharide-induced acute lung injury in mice. Am J Med Sci. 2017;354(5):493–505. doi:10.1016/j.amjms.2017.06.012

81. Yu X, Li C. Protective effects of propofol on experimental neonatal acute lung injury. Mol Med Rep. 2019;19(5):4507–4513. doi:10.3892/mmr.2019.10113

82. Tsaknis G, Siempos II, Kopterides P, et al. Metformin attenuates ventilator-induced lung injury. Crit Care. 2012;16(4):R134. doi:10.1186/cc11439

83. Gao J, Yuan J, Wang Q, et al. Metformin protects against PM2.5-induced lung injury and cardiac dysfunction independent of AMP-activated protein kinase α2. Redox Biol. 2020;28:101345. doi:10.1016/j.redox.2019.101345

84. Wu K, Tian R, Huang J, et al. Metformin alleviated endotoxemia-induced acute lung injury via restoring AMPK-dependent suppression of mTOR. Chem Biol Interact. 2018;291:1–6. doi:10.1016/j.cbi.2018.05.018

85. Hattori Y, Suzuki K, Hattori S, Kasai K. Metformin inhibits cytokine-induced nuclear factor κB activation via AMP-activated protein kinase activation in vascular endothelial cells. Hypertension. 2006;47(6):1183–1188. doi:10.1161/01.HYP.0000221429.94591.72

86. Zhang Y, Zhang H, Li S, Huang K, Jiang L, Wang Y. Metformin alleviates LPS-induced acute lung injury by regulating the SIRT1/NF-κB/NLRP3 pathway and inhibiting endothelial cell pyroptosis. Front Pharmacol. 2022;13:801337. doi:10.3389/fphar.2022.801337

87. Chen H, Ding Y, Chen W, Feng Y, Shi G. Glibenclamide alleviates inflammation in oleic acid model of acute lung injury through NLRP3 inflammasome signaling pathway. DDDT. 2019;13:1545–1554. doi:10.2147/DDDT.S196040

88. rong ZJ, dan LH, Guo C, et al. Berberine attenuates ischemia–reperfusion injury through inhibiting HMGB1 release and NF-κB nuclear translocation. Acta Pharmacol Sin. 2018;39(11):1706–1715. doi:10.1038/s41401-018-0160-1

89. Liu H, You L, Wu J, et al. Berberine suppresses influenza virus-triggered NLRP3 inflammasome activation in macrophages by inducing mitophagy and decreasing mitochondrial ROS. J Leukocyte Biol. 2020;108(1):253–266. doi:10.1002/JLB.3MA0320-358RR

90. Zeng Q, Deng H, Li Y, et al. Berberine directly targets the NEK7 protein to block the NEK7–NLRP3 interaction and exert anti-inflammatory activity. J Med Chem. 2021;64(1):768–781. doi:10.1021/acs.jmedchem.0c01743

91. Chen J, Huang Y, Bian X, He Y. Berberine ameliorates inflammation in acute lung injury via NF-κB/Nlrp3 signaling pathway. Front Nutr. 2022;9:851255. doi:10.3389/fnut.2022.851255

92. Li J, Cao H, Liu P, Cheng G, Sun M. Glycyrrhizic acid in the treatment of liver diseases: literature review. Biomed Res Int. 2014;2014:1–15. doi:10.1155/2014/872139

93. Di Paola R, Menegazzi M, Mazzon E, et al. Protective effects of glycyrrhizin in a gut hypoxia (ischemia)-reoxygenation (reperfusion) model. Intensive Care Med. 2009;35(4):687–697. doi:10.1007/s00134-008-1334-y

94. Wang G, Hiramoto K, Ma N, et al. Glycyrrhizin attenuates carcinogenesis by inhibiting the inflammatory response in a murine model of colorectal cancer. IJMS. 2021;22(5):2609. doi:10.3390/ijms22052609

95. Qu L, Chen C, He W, et al. Glycyrrhizic acid ameliorates LPS-induced acute lung injury by regulating autophagy through the PI3K/AKT/mTOR pathway. Am J Transl Res. 2019;11(4):2042–2055.

96. Wang J, Ren C, Bi W, Batu W. Glycyrrhizin mitigates acute lung injury by inhibiting the NLRP3 inflammasome in vitro and in vivo. J Ethnopharmacol. 2023;303:115948. doi:10.1016/j.jep.2022.115948

97. Wang Q, Xiao L. Isochlorogenic acid A attenuates acute lung injury induced by LPS via Nf-κB/NLRP3 signaling pathway. Am J Transl Res. 2019;11(11):7018–7026.

98. Xuan T, Gong G, Du H, et al. Protective effect of pteryxin on LPS-induced acute lung injury via modulating MAPK/NF-κB pathway and NLRP3 inflammasome activation. J Ethnopharmacol. 2022;286:114924. doi:10.1016/j.jep.2021.114924

99. Han S, Yuan R, Cui Y, et al. Hederasaponin C alleviates lipopolysaccharide-induced acute lung injury in vivo and in vitro through the PIP2/NF-κB/NLRP3 signaling pathway. Front Immunol. 2022;13:846384. doi:10.3389/fimmu.2022.846384

100. Dong Y, Zhang L, Jiang Y, Dai J, Tang L, Liu G. Emodin reactivated autophagy and alleviated inflammatory lung injury in mice with lethal endotoxemia. Exp Anim. 2019; 68(4):559–568. doi:10.1538/expanim.19-0004

101. Liu B, Cheng Y, Wu Y, et al. Emodin improves alveolar hypercoagulation and inhibits pulmonary inflammation in LPS-provoked ARDS in mice via NF-κB inactivation. Int Immunopharmacol. 2020;88:107020. doi:10.1016/j.intimp.2020.107020

102. Liu Y, Shang L, Zhou J, Pan G, Zhou F, Yang S. Emodin attenuates LPS-induced acute lung injury by inhibiting NLRP3 inflammasome-dependent pyroptosis signaling pathway in vitro and in vivo. Inflammation. 2022;45(2):753–767. doi:10.1007/s10753-021-01581-1

103. De La Vega MR, Dodson M, Gross C, et al. Role of Nrf2 and Autophagy in Acute Lung Injury. Curr Pharmacol Rep. 2016;2(2):91–101. doi:10.1007/s40495-016-0053-2

104. Hou Y, Wang Y, He Q, et al. Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury. Behav Brain Res. 2018;336:32–39. doi:10.1016/j.bbr.2017.06.027

105. Liu Y, Zhou J, Luo Y, et al. Honokiol alleviates LPS-induced acute lung injury by inhibiting NLRP3 inflammasome-mediated pyroptosis via Nrf2 activation in vitro and in vivo. Chin Med. 2021;16(1):127. doi:10.1186/s13020-021-00541-z

106. He H, Jiang H, Chen Y, et al. Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity. Nat Commun. 2018;9(1):2550. doi:10.1038/s41467-018-04947-6

107. Yang H, Lv H, Li H, Ci X, Peng L. Oridonin protects LPS-induced acute lung injury by modulating Nrf2-mediated oxidative stress and Nrf2-independent NLRP3 and NF-κB pathways. Cell Commun Signal. 2019;17(1):62. doi:10.1186/s12964-019-0366-y

108. Kang JY, Xu MM, Sun Y, et al. Melatonin attenuates LPS-induced pyroptosis in acute lung injury by inhibiting NLRP3-GSDMD pathway via activating Nrf2/HO-1 signaling axis. Int Immunopharmacol. 2022;109:108782. doi:10.1016/j.intimp.2022.108782

109. Hong H, Lou S, Zheng F, et al. Hydnocarpin D attenuates lipopolysaccharide-induced acute lung injury via MAPK/NF-κB and Keap1/Nrf2/HO-1 pathway. Phytomedicine. 2022;101:154143. doi:10.1016/j.phymed.2022.154143

110. Zhang L, Zhu XZ, Badamjav R, et al. Isoorientin protects lipopolysaccharide-induced acute lung injury in mice via modulating Keap1/Nrf2-HO-1 and NLRP3 inflammasome pathways. Eur J Pharmacol. 2022;917:174748. doi:10.1016/j.ejphar.2022.174748

111. Xue Y, Zhang Y, Chen L, et al. Citrulline protects against LPS‑induced acute lung injury by inhibiting ROS/NLRP3‑dependent pyroptosis and apoptosis via the Nrf2 signaling pathway. Exp Ther Med. 2022;24(4):632. doi:10.3892/etm.2022.11569

112. Kim MJ, Yoon JH, Ryu JH. Mitophagy: a balance regulator of NLRP3 inflammasome activation. BMB Rep. 2016;49(10):529–535. doi:10.5483/BMBRep.2016.49.10.115

113. Che X, Chai J, Fang Y, et al. Sestrin2 in hypoxia and hypoxia-related diseases. Redox Rep. 2021;26(1):111–116. doi:10.1080/13510002.2021.1948774

114. Kim MJ, Bae SH, Ryu JC, et al. SESN2/sestrin2 suppresses sepsis by inducing mitophagy and inhibiting NLRP3 activation in macrophages. Autophagy. 2016;12(8):1272–1291. doi:10.1080/15548627.2016.1183081

115. Wu D, Zhang H, Wu Q, et al. Sestrin 2 protects against LPS-induced acute lung injury by inducing mitophagy in alveolar macrophages. Life Sci. 2021;267:118941. doi:10.1016/j.lfs.2020.118941

116. Berridge MJ, Lipp P, Bootman MD. The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol. 2000;1(1):11–21. doi:10.1038/35036035

117. Murakami T, Ockinger J, Yu J, et al. Critical role for calcium mobilization in activation of the NLRP3 inflammasome. Proc Natl Acad Sci USA. 2012;109(28):11282–11287. doi:10.1073/pnas.1117765109

118. Racioppi L, Means AR. Calcium/calmodulin-dependent kinase IV in immune and inflammatory responses: novel routes for an ancient traveller. Trend Immunol. 2008;29(12):600–607. doi:10.1016/j.it.2008.08.005

119. Zhang T, Li M, Zhao S, et al. CaMK4 promotes acute lung injury through NLRP3 inflammasome activation in type II alveolar epithelial cell. Front Immunol. 2022;13:890710. doi:10.3389/fimmu.2022.890710

120. Berghe TV, Demon D, Bogaert P, et al. Simultaneous targeting of IL-1 and IL-18 is required for protection against inflammatory and septic shock. Am J Respir Crit Care Med. 2014;189(3):282–291. doi:10.1164/rccm.201308-1535OC

121. Selvarani R, Mohammed S, Richardson A. Effect of rapamycin on aging and age-related diseases—past and future. GeroScience. 2021;43(3):1135–1158. doi:10.1007/s11357-020-00274-1

122. Jung CH, Jun CB, Ro SH, et al. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. MBoC. 2009;20(7):1992–2003. doi:10.1091/mbc.e08-12-1249

123. Chuang SY, Yang CH, Chou CC, Chiang YP, Chuang TH, Hsu LC. TLR-induced PAI-2 expression suppresses IL-1β processing via increasing autophagy and NLRP3 degradation. Proc Natl Acad Sci USA. 2013;110(40):16079–16084. doi:10.1073/pnas.1306556110

124. Nakahira K, Haspel JA, Rathinam VAK, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12(3):222–230. doi:10.1038/ni.1980

125. Hu Y, Lou J, Mao YY, et al. Activation of MTOR in pulmonary epithelium promotes LPS-induced acute lung injury. Autophagy. 2016;12(12):2286–2299. doi:10.1080/15548627.2016.1230584

126. Lorne E, Zhao X, Zmijewski JW, et al. Participation of mammalian target of rapamycin complex 1 in toll-like receptor 2– and 4–induced neutrophil activation and acute lung injury. Am J Respir Cell Mol Biol. 2009;41(2):237–245. doi:10.1165/rcmb.2008-0290OC

127. Jia X, Cao B, An Y, Zhang X, Wang C. Rapamycin ameliorates lipopolysaccharide-induced acute lung injury by inhibiting IL-1β and IL-18 production. Int Immunopharmacol. 2019;67:211–219. doi:10.1016/j.intimp.2018.12.017

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.