Abstract
Oxidative stress is an important contributor to sepsis and one of the most important causes of death in intensive care units. Even though sepsis pathogenesis remains obscure due to its heterogenicity and complexity, there is increasing evidence that oxidants and antioxidants play a key role in its onset and progression. Recent evidence suggests that pyroptosis is required for defense against bacterial infection, and it is active in several cell types during sepsis. One of the most relevant mediators of pyroptosis is the NLRP3 inflammasome, and the oxidative stress/NLRP3 signaling pathway is one of the main upstream signals involved in its activation. So, it is of special relevance to clarify how oxidative stress and antioxidants can modulate pyroptosis signals and therefore decrease the deleterious effects that both oxidative stress and pyroptosis-related cytokines can induce in the tissues during sepsis. Recent studies evaluating several antioxidants are promising, but further trials are needed to confirm their potential role as agents to block NLRP3 and mitigate the exacerbated inflammasome-related responses during sepsis.
Introduction
Sepsis is a life-threatening condition caused by an abnormal host response to an infection that produces altered physiological responses damaging patients’ own tissues and results in organ dysfunction and death (Singer et al. 2016).
Sepsis is the leading cause of mortality in the intensive care units (ICU) (Angus et al. 2001). Specifically, the latest epidemiologic studies estimate almost 50 million cases of sepsis worldwide and up to 10 million deaths every year (Rudd et al. 2020, Beltrán-García et al. 2022).
Clinically, sepsis is characterized by a simultaneous hyperinflammatory and anti-inflammatory response, which contribute to the deregulation of the patient’s immune system (Nakamori et al. 2020, Lazzaro et al. 2022). Moreover, circulatory changes, cardiovascular problems (Beltrán-García et al. 2021), neurological deficits (Osca-Verdegal et al. 2021), mitochondrial dysfunction and impaired respiration (Mantzarlis et al. 2017) occur in septic patients, increasing the risk of death.
Regarding the deregulation of the immune system during sepsis pathophysiology, pyroptosis has been proposed as one of the mechanisms that mediate sepsis immune dysregulation (Nakamori et al. 2020). Specifically, pyroptosis is defined as a mechanism of programmed cell death that mediates inflammation (Nakamori et al. 2020). In recent years, it has been demonstrated that this type of cell death occurs in many human cells, affecting the immune system (Boise & Collins 2001), CNS (Liu et al. 1999) and cardiovascular system (Yue et al. 2021), among others, highlighting the importance of this mechanism in a large number of biological systems. Likewise, pyroptosis has been described as a key mediator of a wide range of diseases, including sepsis (Gao et al. 2018). The reason is that pyroptosis plays a fundamental role in regulating the early immune response against numerous infections, including bacterial, viral, protozoan or fungal infections (Man et al. 2017).
Pyroptosis is triggered by various stimuli, such as changes in membrane potential, oxidative stress, hyperglycemia and even inflammation itself (Lamkanfi & Dixit 2011) (Fig. 1). Among them, oxidative stress has been postulated as a central mediator of pyroptosis, playing a critical role in sepsis (Yang et al. 2019). Recent sepsis guidelines (SEPSIS-3) do not consider oxidative stress as an important contributor to sepsis pathophysiology (Singer et al. 2016). Nonetheless, despite the increasing evidence of the role that oxidative stress is playing in sepsis, the implications of oxidative stress in the activation of pyroptosis have been little explored in sepsis.
The aim of this review is to point out the role of oxidative stress in the modulation of pyroptosis and therefore the immune system during sepsis, as well as their role as an effective goal for therapy in sepsis disease.
Inflammasome activation is mediated by oxidative stress
The immune system can be activated in response to pattern recognition receptors (PRRs), which are derived from pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) produced from exogenous and endogenous mediators, respectively (Kang et al. 2015), triggering downstream inflammatory pathways to eliminate microbial infection and repair damaged tissues (Christgen et al. 2020).
Inflammasomes are a group of intracellular multimeric protein complexes that activate inflammatory caspase-1 by the cleavage of pro-caspase-1 (Franchi et al. 2009). Inflammasome’s activation is a major inflammatory pathway and as being part of innate immunity, it can be induced by infectious agents and also by sepsis (Franchi et al. 2009, Christgen et al. 2020). Some inflammasomes are activated by AMPs and DAMPs, while others are activated by cell homeostatic environment modifications caused by those patterns (Christgen et al. 2020). The sensor protein (PRR) characterizes the inflammasome. Five different PRR have been described to form inflammasomes: the nucleotide-binding oligomerization domain (NOD), absent-in-melanoma 2 (AIM2), leucine-rich repeat (LRR)-containing proteins, nucleotide-binding oligomerization domain-like receptor (NLR) family members NLRP1, NLRP3 and NLRC4, in addition to pyrin (Lamkanfi & Dixit 2014, Sharma & Kanneganti 2016).
Among the different PRRs, NLR family members and particularly NLRP3 stands out over others since it is the most extensively studied inflammasome. NLRP3 is a crucial protein participating in the innate immune response to infection (Sutterwala et al. 2014). The most probable reason for the widespread interest in NLRP3 above other NLRs is because it is activated by several stimuli such as extracellular ATP, biological particulate matter and pore-forming toxins and pathogens, including bacteria, viruses and fungi (Mariathasan et al. 2006, Martinon et al. 2006, Li et al. 2008, Gross et al. 2009, Yazdi et al. 2010, Wang et al. 2014). Importantly, NLRP3 was the only one activated by oxidative stress (Sharma et al. 2018). In this regard, Lucena Lage et al. demonstrated recently a direct relationship between lipid peroxidation and NLRP3 activation, broadening the several activators of NLRP3 as a consequence of increased oxidative stress (Lage et al. 2021).
Specifically, NLRP3 inflammasome is formed by different subunits: the NLR family, pyrin domain (PYD)-containing 3 (NLRP3) and an adapter molecule, apoptosis-associated speck-like protein containing a CARD (ASC), which is an adapter that connects the PRR with the downstream effector, caspase-1 (CASP1) (Christgen et al. 2020). After the activation of CASP1, it processes the proinflammatory cytokines interleukin-1β (IL-1β) and IL-18 into their mature forms (Thornberry et al. 1992). Moreover, CASP1 is also responsible for cleaving gasdermin D (GSDMD) protein, which oligomerizes to form pores in the cell membrane, allowing the proinflammatory cytokines to leak out of the cell, thus completing pyroptotic cell death (Yaron et al. 2015, Liu et al. 2016, Sborgi et al. 2016, Ding et al. 2016) (Fig. 1). Activation of NLRP3 inflammasome enables several immune cells such as monocytes and macrophages to release high levels of interleukin IL-1α and IL-1β, amplifying the inflammatory response, as well as IL-18, which modulates the initial immune response during sepsis (Beltrán-García et al. 2022).
NLRP3 upstream regulatory molecules
The upstream regulatory molecules participating in NLRP3 inflammasome activation have not been yet well characterized. It is unknown if NLRP3 is activated directly by a specific ligand through indirect signals or even through both mechanisms. Many direct stimuli have been described as NLRP3 inflammasome activators, such as ATP, K+ and Ca2+ dysregulation, lysosomal disruption by crystalline particulates, mitochondrial DNA (mtDNA) and ROS (Mariathasan et al. 2006, Dostert et al. 2008, Hornung et al. 2008, Yaron et al. 2015, Zhong et al. 2018). Owing to the high number of molecules able to activate the NLRP3 inflammasome, and the chemical and structural differences existing between some of the molecules inducing inflammasome activation, it is expected that there is not a single ligand activating this inflammasome. A new study has described a protein necessary for NLRP3 activation, so it is feasible to think that NLRP3 inflammasome is activated by indirect pathways (Samir et al. 2019). Specifically, the authors found that DDX3X, which is sequestered by stress granules found in cytoplasmic compartments, helps cells survive different stressors (Protter & Parker 2016). Since DDX3X drives inflammasome activation, NLRP3 inflammasome and stress granules compete for DDX3X molecules to orchestrate the innate responses activation and subsequent cell-fate decisions under stress conditions (Samir et al. 2019).
Although upstream pathways are not well elucidated (Yuk et al. 2020), it was shown that K+ efflux activates NLRP3 in response to a high amount of stimuli, including membrane pores formed by toxins, and has been proposed to be a common mechanism of NLRP3 activation (Pétrilli et al. 2007, Muñoz-Planillo et al. 2013). In fact, trans-golgi network (TGN) disturbance has been related to the activation of NLRP3 through K+-dependent and K+-independent NLRP3 activators (Chen & Chen 2018). Nevertheless, NLRP3 inflammasome can be activated also by molecules that are K+ efflux independent, such as those small molecules that target the mitochondria (Groß et al. 2016), which release ROS and mtDNA. Importantly, both ROS and mtDNA have been involved in NLRP3 inflammasome activation, emphasizing the mitochondrial relevance for NLRP3 activation (Dostert et al. 2008, Misawa et al. 2013, Liu et al. 2018, Zhong et al. 2018).
Occasionally, NLRP3 levels within the cell are not enough to generate a strong inflammasome activation. Therefore, the current NLRP3 canonical activation model proposes a two-step process. The first signal guides the upregulation of NLRP3 inflammasome components, at transcriptional and post-translational levels, facilitating its activation (Sutterwala et al. 2014, Christgen et al. 2020). After priming, a later unchain signal is required. In this scenario, NLRP3 oligomerizes and binds to ASC, and inflammasome activation continues (Sutterwala et al. 2014, Christgen et al. 2020) (Fig. 1).
NLRP3 activation mediated by ionic imbalance
There are many DAMPs and PAMPs that cause an efflux of K+ activating NLRP3 inflammasome. These molecules can cause a K+ efflux by direct pathways, such as nigericin, that forms pores on plasma membranes, or by indirect pathways, such as ATP-induced NLRP3 inflammasome activation, that is activated by the passage of ATP through P2RX7, a cation channel that works as a receptor of extracellular ATP (Gustin et al. 2015, Di et al. 2018, Gong et al. 2018, Hafner-Bratkovič & Pelegrín 2018). However, potassium efflux has also been found to be mediated by GSDMD (Shi et al. 2015, Orning et al. 2018, Sarhan et al. 2018, Chen et al. 2019).
One of the major events that modulate the activation of NLRP3 is changes in the membrane potential. In fact, several authors demonstrated that damaging the plasmatic membrane could increase the levels of NLRP3 (Beckwith et al. 2020, Paik et al. 2021), and this activation is usually mediated by changes in the membrane potential led by different ion concentrations (Fig. 2).
In this regard, potassium has a prominent role in modulating the activation of NLRP3 (Muñoz-Planillo et al. 2013, Gong et al. 2018), since other inflammasomes’ activation is not affected by the concentration of K+. In fact, lower concentration of cytosolic K+ has been related to induction of NLRP3 activation and high concentration of K+ has been associated with an inhibition of NLRP3 inflammasome activation (Franchi et al. 2007, Pétrilli et al. 2007, Muñoz-Planillo 2013).
Similarly, numerous studies indicated that NLRP3 inflammasome requires calcium influx or mobilization for its activation (Compan et al. 2012, Lee et al. 2012, Murakami et al. 2012, Zhong et al. 2013, Yaron et al. 2015). Nigericin, one of the best-characterized pyroptosis NLRP3-specific inducers, works by causing plasma membrane pores that allows the Ca2+ in-flow, therefore activating NLRP3 inflammasome (Murakami et al. 2012). Likewise, Brough et al. demonstrated that when Ca2+ is chelated, the inflammasome activation is inhibited (Brough et al. 2003). Other authors also demonstrated how the endoplasmic reticulum (intracellular Ca2+ reservoir) plays a critical role in modulating the activation of NLRP3 (Lee et al. 2012). Increased levels of extracellular Ca2+ induce the NLRP3 activation by stimulating G protein-coupled calcium-sensing receptors and GPRC6A and activation of the phosphatidyl inositol/Ca(2+) pathway (Rossol et al. 2012).
Additionally, other ions like Cl− have been related to NLRP3 inflammasome activation. Verhoef and colleagues reported a decrease in intracellular Cl−-induced IL-1β secretion (Verhoef et al. 2005). In this regard, Cl− channel blockers such as flufenamic acid, mefenamic acid and benzoic acid, among others, have been demonstrated to be able to inhibit NLRP3 inflammasome (Channappanavar et al. 2016, Daniels et al. 2016, Tang et al. 2017). Specifically, they found that a reduction in intracellular Cl− concentration leads to an increase in cation flow hence producing the activation of NLRP3. Moreover, other authors also demonstrated that chloride intracellular channels act downstream of the potassium efflux–mitochondrial reactive oxygen species (ROS) axis to promote NLRP3 inflammasome activation (Daniels et al. 2016, Tang et al. 2017). These findings demonstrate that Cl− efflux and influx play an important role in NLRP3 inflammasome activation and could be even more important in neuroinflammation, as a consequence of GABA receptors activation that modulates NLRP3 activation through changes in intracellular Cl− (Lang et al. 2020, Omer et al. 2020).
Comparable to K+, Ca2+ and Cl−, Na+ is other molecule that is involved in NLRP3 activation (Muñoz-Planillo et al. 2013). Na+ influx activates NLRP3 inflammasome by reducing the decrease in intracellular K+ (Schorn et al. 2011). So, Na+ influx can modulate the activation of NLRP3 by decreasing intracellular K+. Moreover, other ions such as Zn2+ have also been proposed to induce the activation of NLRP3 in some immune cells such as macrophages (Summersgill et al. 2014), because zinc depletion damages the integrity of the lysosomal membrane, inducing a release of pyroptosis-related cytokines (Summersgill et al. 2014).
Mitophagy and mitochondrial oxidative stress
Autophagy is defined as a non-selective degradation process for intracellular cargos (Yuk et al. 2020), but it has been also described as a selective process that is able to target some organelles and substances (Khaminets et al. 2016). Particularly, mitophagy has been described as a selective autophagy process that is able to target and destroy the mitochondria for maintaining its homeostasis, through regulation of mitochondrial ROS generation and elimination of damaged mitochondria (Ashrafi & Schwarz 2013, Wei et al. 2015, Hamacher-Brady & Brady 2016, Gustafsson & Dorn 2019). Mitophagy dysregulation causes a cascade of reactions, including mitochondrial dysfunction, increased production of mitochondrial ROS and translocation of mtDNA into the cytosol, leading to the inflammasome’s activation, mitochondrial dysfunction, increased production of mitochondrial ROS and translocation of mtDNA into the cytosol (Zhao et al. 2018, Fan et al. 2019, AA et al. 2020), since mtDNA acts as a crucial DAMP signal to activate NLRP3 inflammasome (Elliot & Sutterwala 2015, Jin et al. 2017, Bae et al. 2019). In this regard, defective mitophagy leads to mitochondrial dysfunction and impairment of mitochondrial membrane integrity, opening mitochondrial permeability transition pore (mPTP) which causes an mtDNA leak from damaged mitochondria, thus forming a vicious cycle that ultimately contributes to inflammation (Harrington et al. 2017, Rottenberg & Hoek 2017, Xu et al. 2020) (Fig. 2). This is of special interest since defective mitophagy causes inflammasome activation, which in turn contributes to the pathogenesis of many human diseases, including sepsis (Harris et al. 2018, Kim et al. 2016, Wu et al. 2019).
As previously described, the inflammasome is activated as a consequence of mitochondrial perturbation and dysfunction. It is noteworthy that the NLRP3 inflammasome is highly active due to an overload of mitochondrial Ca2+ that increases the generation of mitochondrial ROS (Tschopp 2011, Abais et al. 2015, Elliot & Sutterwala 2015, Yu & Lee 2016).
Mitochondrial DAMPs (mtDAMPs) have gained relevance among researchers in the last years. They include mtDNA, mitochondrial transcription factor (TFAm), cardiolipin and ATP (Khwaja et al. 2021). These molecules are released during tissue injury and activate a similar immune response to that caused by pathogens (Kang et al. 2015). Among them, probably the most important molecule in this process is mtDNA, whose release causes the activation of NLRP3 inflammasome through TLR9-NF-κβ pathway (Shimada et al. 2012, West & Shadel 2017, Khwaja et al. 2021). The mtDNA can be leaked into the cytosol easily when the mitochondrial membrane potential is decreased as a consequence of mitochondrial stress (Harrington et al. 2017, Liu et al. 2018). Recently, the relationship between mtDNA, mitochondrial ROS and NLRP3 has been described (Sok et al. 2021, Xian et al. 2021). Sok et al. observed that by inhibiting the release of oxidized mitochondrial DNA, the mitochondrial ROS generation was decreased and subsequently, NLRP3 inflammasomes were inhibited (Sok et al. 2021). Wu et al. (2020) performed several experiments inducing delayed resuscitation in rats and showed that delayed resuscitation caused liver damage and oxidative stress, developing a liver injury through mitochondrial destruction and mtDNA/NLRP3 axis activation. Moreover, they also observed that the structure and function of mitochondria were protected by antioxidants directed to inhibit the release of mtDNA (Wu et al. 2020). These results denoted that mtDNA has a direct role in the development of systemic inflammation and organ dysfunction, via the NLRP3 inflammasome. Interestingly, some studies have correlated the plasmatic levels of mtDNA with the septic shock clinical outcome (Jiménez-Sousa et al. 2015, Hu et al. 2017, Schneck et al. 2020) and have proposed mtDNA as a biomarker to predict mortality in patients with sepsis better than lactate concentration or sequential organ failure assessment score (SOFA) (Kung et al. 2012). Since elevated circulating levels of mtDNA have been directly linked to the activation of NLRP3 inflammasome, it is plausible to use the circulating mtDNA levels as a biomarker for sepsis severity and prognosis.
Evidence supporting the relevance of oxidative stress in sepsis therapies
NLRP3 has been postulated as an important mediator of sepsis (Cassel & Sutterwala 2010, Kalbitz et al. 2016, Zhang et al. 2019), and some therapeutic strategies under research are directed to block NLRP3 overactivation, demonstrating their important role in sepsis development and pathogenesis (Danielski et al. 2020, Zhang & Ning 2021).
In recent years, several studies have postulated that sepsis pathogenesis is affected by oxygen metabolism accelerating the disease progression and causing multiple organ failure (Kozlov & Grillari 2022). Similarly, the cGMP-dependent protein kinase 1 alpha (PRKG1) activation by oxidative stress induces a blood vessel dilatation, increasing permeability and therefore decreasing cardiac output (Banday & Lokhandwala 2019) and organ perfusion as well as oxygen delivery, inducing ischemia to organs and finally organ failure (Rudyk et al. 2013). In addition, excessive oxidative stress is a common feature of sepsis, mainly the disruption of the redox homeostasis (Lopes-Pires et al. 2021). For example, some reactive species found in sepsis pathogenesis include superoxide (O2 ·−), hydroxyl radical (·OH), hydrogen peroxide (H2O2), peroxynitrite (ONOO·−) and hypochlorous acid (HOCl) (Parihar et al. 2008). Conversely, levels of some antioxidants such as α-tocopherol (Takeda et al. 1984, Ogilvie et al. 1991), selenium (Mishra et al. 2007, Sakr et al. 2007), ascorbic acid (Borrelli et al. 1996), reduced glutathione (GSH) (Brealey et al. 2002), vitamin A, β-carotene and lycopene (Rossol et al. 2012) have been found to be decreased in septic patients. Consequently, therapies targeting redox abnormalities have been postulated as promising strategy for improving the management and treatment of septic patients.
Several antioxidants have been studied in animal models as well as in clinical trials to develop effective therapies against sepsis. In this regard, Escames et al. tested melatonin in a mice model of sepsis (cecal ligation puncture (CLP) sepsis model) (Escames et al. 2007). They observed that sepsis increased the expression levels of iNOS and mitochondrial NOS, accompanied by an increase in oxidative stress and reduced ATP production and respiratory chain impairment, despite ATPase levels did not change. In addition, mitochondrial NOS induction was associated with heart mitochondrial dysfunction. The treatment with the antioxidant and circadian regulator melatonin was able to reduce iNOS and mitochondrial NOS expression levels avoiding the mitochondrial homeostasis impairment in sepsis mice and reestablishing mitochondrial ATP production (Escames et al. 2007).
Similarly, in a CLP sepsis study in rats, sepsis induced a decrease in GSH concentrations while producing an increase in malondialdehyde (MDA) levels and myeloperoxidase (MPO) activity (neutrophil aggregation) in the evaluated tissues (liver, lung, heart, kidney and brain). Likewise, sepsis also led to higher plasma aspartate aminotransferase (AST) and alanine transaminase (ALT) activity, as well as creatinine (Cre) levels and blood urea nitrogen. Treatment with melatonin (10 mg/kg i.p.) before and after the surgery restored the MDA levels in the evaluated tissues and restored GSH levels in kidney, diaphragm and brain. In cardiac and pulmonary tissues, no improvement was reported Moreover, melatonin treatment reduced MPO activity in all tissues and returned to normal levels of AST, ALT, blood urea nitrogen (BUN) and Cre levels (Zhang et al. 2017). These findings highlight the potential of melatonin as therapy or co-therapy (supporting therapy) for sepsis disease.
Another promising antioxidant in sepsis is N-acetylcysteine (NAC). Some studies demonstrated that lipopolysaccharide (LPS) injections into mice (LPS sepsis model) increase BUN, LDH, Cre and CPK levels, as well as other pro- and anti-inflammatory cytokines such as IL-1β, TNF-α, IL-6 and IL-10 levels. The mean arterial blood pressure (MAP) was found to be decreased after exposition to LPS mice treatments, which represents a serious risk of death and end-organ failure. Conversely, treatment with NAC moderately avoids the decrease in MAP and markers of organ injury (BUN, LDH, Cre and CPK) and pro- and anti-inflammatory biomarkers (IL-1β, TNF-a, IL-6, and IL-10) caused by LPS injection (sepsis) (Hsu et al. 2006).
The work by Blackwell and colleagues supports the idea of the use of NAC as potential treatment against sepsis (Blackwell et al. 1996). In this work, authors administered NAC before injecting endotoxin to rats (sepsis animal model) demonstrating that NAC reduced NF-kB activation in a dose-dependent manner, reducing the cytokine-induced neutrophil chemoattractant expression in lung tissue (Blackwell et al. 1996). Since NF-kB is a master regulator of NLRP3 and pyroptosis (Cornut et al. 2020), NAC treatment, through downregulating NF-kB activation, demonstrated to be able to diminish the damage and the inflammatory response, hence alleviating several symptoms of sepsis typically associated to respiratory distress (Blackwell et al. 1996).
Other well-known antioxidants are vitamin C and vitamin E. Both vitamins have demonstrated their therapeutic potential against sepsis in in vivo studies. For example, recently, in a study carried on by Marik et al., vitamin C reduced the elevation of aminotransferase and hepatic lipid peroxide and MDA levels in a CLP sepsis rat model. Moreover, hepatic GSH concentration was higher in the sepsis group, after vitamin C treatment. In addition, vitamin C also prevented the expression of the vasodilatory gene ET1 and its receptor ETB in the liver. Finally, the increased levels of iNOS and heme oxygenase-1 found in septic rats were also attenuated by vitamin C (Kim & Lee 2004).
Similarly, in another study, it was demonstrated that not only vitamin C but also vitamin E treatment reduced the increase of aminotransferase and MDA during sepsis. Similar to the previous study, the decreased expression levels of hepatic CYP1A1 and CYP2E1 found in rat sepsis model were prevented by both vitamins, vitamins C and E (Kim & Lee 2006). Since elevated levels of aminotransferase and MDA have been related to elevated levels of NLRP3 (Wu et al. 2017, Lin et al. 2018, Chen et al. 2020), it is plausible that antioxidant combinatory therapy could modulate inflammasome activation and therefore prevent the hyperinflammatory response in septic patients.
Regarding therapies already being applied in sepsis, Manzanares and colleagues performed a meta-analysis study on the effects of antioxidant co-therapy in septic patients based on 21 clinical trials (Manzanares et al. 2012). They found that the antioxidant combinatory therapy induced a reduction of the mechanical ventilation need and a decrease in infections, although there was no effect related to ICU or hospital length of stay. Furthermore, the use of combined antioxidants was correlated with a mortality reduction in septic patients with elevated risk of death (Manzanares et al. 2012).
From different clinical studies performed so far, selenium, used as co-adjuvant antioxidant therapy, demonstrated to be one of the most feasible candidates to be used as a therapy, since its use showed decreased mortality in critically ill patients (Hardy et al. 2012, Huang et al. 2013). Specifically, in sepsis, clinical studies performed on septic patients demonstrated that selenium supplementation increased glutathione peroxidase activity, as well as a significant negative correlation between plasmatic selenium and SOFA score (Mishra et al. 2007). Similarly, another clinical study performed on critically ill patients in ICUs reported that selenium supplementation reduced renal failure and improved patient outcomes (Angstwurm et al. 1999).
Several clinical studies, applying antioxidant molecules as therapy or co-adjuvant therapy against sepsis, have been developed, with the aim of ameliorating the effects of oxidative stress in septic patients. So far, 47 clinical trials have been registered on clinicaltrials.gov, focused to mitigate the damage mediated by oxidative stress in sepsis. The strategies developed to date are diverse, from the use of cellular antioxidants such as selenium (NCT00207844) or glutamine (NCT00133978, NCT01367223) to vitamin C and vitamin D (NCT04216459) or vitamin B1 (NCT03380507), some molecules with antioxidant activity as melatonin (NCT03295162, NCT01724424) and other strategies focused on decreasing oxidative damage as probiotic-based dietary supplement (NCT01474629), specific molecules as acetaminophen (NCT01739361), dopexamine, epinephrine and norepinephrine (NCT00134212), deferasirox (NCT01349699) or atazanavir (NCT00916448), or even some medical strategies such as polymyxin B hemoperfusion (NCT01756755). Nevertheless, despite several of these therapies having already demonstrated improved clinical outcomes in septic patients, many of these proposed therapies have yet demonstrated enough efficacy to be used as a sepsis treatment alone. Further studies are needed in order to demonstrate its potential as co-adjuvant agent to be used in a combinatory therapy against sepsis (see Table 1).
Sepsis therapies have been tested currently in clinical trials modulating oxidative stress levels.
Drug/therapies | NCT | Recruitment status | Type of study | Status | Sponsor |
---|---|---|---|---|---|
Selenium | NCT00207844 | Completed | Interventional randomized |
Ph. 2 | Centre Hospitalier de Meaux, Ministry of Health, France |
Glutamine | NCT00133978 | Completed | Interventional randomized |
N.A. | Clinical Evaluation Research Unit at Kingston General Hospital, Canada |
NCT01367223 | Completed | Interventional randomized |
Ph. 4 | Fundació Sant Joan de Déu, Spanish National Health System, Spain | |
Vitamin C and vitamin D | NCT04216459 | Completed | Interventional randomized |
N.A. | Mansoura University, Egypt |
Vitamin B1 | NCT03380507 | Completed | Interventional randomized |
Ph.2-Ph.3 | Hamad Medical Corporation, Qatar |
Melatonin | NCT03295162 | Completed | Interventional randomized |
Ph.1-Ph.2 | Ain Shams University, Egypt |
NCT01724424 | Completed | Interventional non-randomized |
Ph.1 | University of Aberdeen, Scotland, UK | |
Acetaminophen | NCT01739361 | Completed | Interventional randomized |
Ph. 2 | Vanderbilt University, Tennessee, USA |
Dopexamine, epinephrine and norepinephrine | NCT00134212 | Completed | Interventional randomized |
Ph. 2 | Rennes University Hospital, France |
Deferasirox | NCT01349699 | Completed | Interventional randomized |
N.A. | Radboud University Medical Center, Netherlands |
Atazanavir | NCT00916448 | Completed | Interventional randomized |
N.A. | Radboud University Medical Center, Netherlands |
Polymyxin B hemoperfusion | NCT01756755 | Completed | Interventional randomized |
N.A. | National Taiwan University Hospital, Taiwan |
N.A., not applicable; Ph, clinical phase.
Conclusions
It is broadly accepted that oxidative stress plays an important role in the pathogenesis of sepsis (Prauchner et al. 2017). In physiological conditions, an equilibrium between oxidant and antioxidant systems inside the cell usually occurs. Oxidative stress is an important contributor to sepsis physiopathology (Mantzarlis et al. 2017). Continuous oxidative stress leads to an increase in the inflammatory response, inducing mitochondria alterations, which in turn can lead to mitochondrial dysfunction and finally cell death, mediating sepsis-induced organ failure (Exline & Crouser 2008).
It is noteworthy, that during an inflammatory process, the immune system produces a huge amount of oxidant species in a process called respiratory burst, which contributes to the immune defense against invading pathogens. However, ROS produced during the respiratory burst can also damage cellular components and interestingly can act as signals to modulate the initiation of pyroptosis, through the activation of NLRP3, since it is the only type of inflammasome activated by oxidative stress. Furthermore, in recent years, the notion that sepsis pathogenesis also involves an inability of the cell to consume oxygen has increased. Because consumption of molecular oxygen by mitochondria accounts for 98% of the body’s O2 usage (Singer 2014), it is expected that problems in O2 consumption contribute to mitochondrial dysfunction and play an important role in the pathogenesis of sepsis (Singer 2014, Rahmel et al. 2020). Despite sepsis being a heterogeneous and complex condition, oxidative stress is a common feature of sepsis, so therapies focused on reducing the redox changes may be useful to improve the management of septic patients, specifically therapies directed to modulate the NLRP3 activation, which subsequently attenuates the inflammatory response during sepsis (Zhang et al. 2018). Unfortunately, a whole understanding of the mechanisms leading to oxidative stress in sepsis progression is not currently available; however, it is well-established that an energy deficit in cells by mitochondrial dysfunction and impaired vascular tone are central events that lead to multi-organ dysfunction and therefore poorer clinical outcomes and increased mortality in septic patients (Mantzarlis et al. 2017).
In this regard, numerous trials have been performed to identify potential treatments against sepsis over the years. Particularly, antioxidants targeting mitochondria stand out among them, since they can accumulate inside the inner membrane of the mitochondria, and therefore, they can modulate different molecular mechanisms such as pyroptosis, inflammation and cellular respiration, among other molecular events that occur during sepsis. Nevertheless, the results obtained in clinical trials are not sufficient for their implementation in sepsis management, and further research is still needed, especially to understand the molecular mechanisms that underlie oxidative stress during sepsis physiopathology and its contribution to NLRP3 inflammasome activity.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.
Funding
J B-G is supported by Contratos i-PFIS grant (IFI18/00015) and co-financed by the European Social Fund. R O-V is supported by Contratos PFIS grant (FI20/00202) from AES-ISCIII and co-financed by the European Regional Development Fund (ERDF). J L G-G thanks INCLIVA and GVA for the starting grants (GV/2014/132) and AES2016 (ISCIII) for grant numbers PI16/01036 and PI19/00994, co-financed by the European Regional Development Fund (ERDF). J L G-G and F V P thank Grand Challenges Canada and the Spanish Ministry of Economy and Competitiveness, ISCIII through CIBERer (Biomedical Network Research Center for Rare Diseases and INGENIO2010). We would like to thank INCLIVA’s Biobank and MEC, ISCIII, FEDER for grant PT13/0010/0004.
Author contribution statement
J B-G and R O-V: manuscript drafting. F V P and J L G-G: critical revision. All the authors read and approved the final manuscript.
References
Abais JM, Xia M, Zhang Y, Boini KM & Li PL 2015 Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxidants and Redox Signaling 22 1111–1129. (https://doi.org/10.1089/ars.2014.5994)
AA MA, Ameenudeen S, Kumar A, Hemalatha S, Ahmed N, Ali N, AlAsmari AF, Aashique M & Waseem M 2020 Emerging role of mitophagy in inflammatory diseases: cellular and molecular episodes. Current Pharmaceutical Design 26 485–491. (https://doi.org/10.2174/1381612826666200107144810)
Angstwurm MWA, Schottdorf J, Schopohl J & Gaertner R 1999 Selenium replacement in patients with severe systemic inflammatory response syndrome improves clinical outcome. Critical Care Medicine 27 1807–1813. (https://doi.org/10.1097/00003246-199909000-00017)
Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J & Pinsky MR 2001 Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Critical Care Medicine 29 1303–1310. (https://doi.org/10.1097/00003246-200107000-00002)
Ashrafi G & Schwarz TL 2013 The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death and Differentiation 20 31–42. (https://doi.org/10.1038/cdd.2012.81)
Bae JH, Jo SI, Kim SJ, Lee JM, Jeong JH, Kang JS, Cho NJ, Kim SS, Lee EY & Moon JS 2019 Circulating cell-free mtDNA contributes to AIM2 inflammasome-mediated chronic inflammation in patients with type 2 diabetes. Cells 8 328. (https://doi.org/10.3390/cells8040328)
Banday AA & Lokhandwala MF 2019 Oxidative stress impairs cGMP-dependent protein kinase activation and vasodilator-stimulated phosphoprotein serine-phosphorylation. Clinical and Experimental Hypertension 41 5–13. (https://doi.org/10.1080/10641963.2018.1433197)
Beckwith KS, Beckwith MS, Ullmann S, Sætra RS, Kim H, Marstad A, Åsberg SE, Strand TA, Haug M & Niederweis M et al.2020 Plasma membrane damage causes NLRP3 activation and pyroptosis during Mycobacterium tuberculosis infection. Nature Communications 11 2270. (https://doi.org/10.1038/s41467-020-16143-6)
Beltrán-García J, Osca-Verdegal R, Nácher-Sendra E, Cardona-Monzonís A, Sanchis-Gomar F, Carbonell N, Pallardó FV, Lavie CJ & García-Giménez JL 2021 Role of non-coding RNAs as biomarkers of deleterious cardiovascular effects in sepsis. Progress in Cardiovascular Diseases 68 70–77. (https://doi.org/10.1016/j.pcad.2021.07.005)
Beltrán-García J, Osca-Verdegal R, Jávega B, Herrera G, O’Connor JE, García-López E, Casabó-Vallés G, Rodriguez-Gimillo M, Ferreres J & Carbonell N et al.2022 Characterization of early peripheral immune responses in patients with sepsis and septic shock. Biomedicines 10 525. (https://doi.org/10.3390/biomedicines10030525)
Blackwell TS, Blackwell TR, Holden EP, Christman BW & Christman JW 1996 In vivo antioxidant treatment suppresses nuclear factor-κ B activation and neutrophilic lung inflammation. Journal of Immunology 157 1630–1637. (PMID: https://pubmed.ncbi.nlm.nih.gov/8759749/)
Boise LH & Collins CM 2001 Salmonella-induced cell death: apoptosis, necrosis or programmed cell death? Trends in Microbiology 9 64–67. (https://doi.org/10.1016/S0966-842X(0001937-5)
Borrelli E, Roux-Lombard P, Grau GE, Girardin E, Ricou B, Dayer J & Suter PM 1996 Plasma concentrations of cytokines, their soluble receptors, and antioxidant vitamins can predict the development of multiple organ failure in patients at risk. Critical Care Medicine 24 392–397. (https://doi.org/10.1097/00003246-199603000-00006)
Brealey D, Brand M, Hargreaves I, Heales S, Land J, Smolenski R, Davies NA, Cooper CE & Singer M 2002 Association between mitochondrial dysfunction and severity and outcome of septic shock. Lancet 360 219–223. (https://doi.org/10.1016/S0140-6736(0209459-X)
Brough D, Le Feuvre RA, Wheeler RD, Solovyova N, Hilfiker S, Rothwell NJ & Verkhratsky A 2003 Ca2+ stores and Ca2+ entry differentially contribute to the release of IL-1 β and IL-1 α from murine macrophages. Journal of Immunology 170 3029–3036. (https://doi.org/10.4049/jimmunol.170.6.3029)
Cassel SL & Sutterwala FS 2010 Sterile inflammatory responses mediated by the NLRP3 inflammasome. European Journal of Immunology 40 607–611. (https://doi.org/10.1002/eji.200940207)
Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK & Perlman S 2016 Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host and Microbe 19 181–193. (https://doi.org/10.1016/j.chom.2016.01.007)
Chen J & Chen ZJ 2018 PtdIns4P on dispersed trans-Golgi network mediates NLRP3 inflammasome activation. Nature 564 71–76. (https://doi.org/10.1038/s41586-018-0761-3)
Chen KW, Demarco B, Heilig R, Shkarina K, Boettcher A, Farady CJ, Pelczar P & Broz P 2019 Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. EMBO Journal 38 e101638. (https://doi.org/10.15252/embj.2019101638)
Chen Y, Que R, Lin L, Shen Y, Liu J & Li Y 2020 Inhibition of oxidative stress and NLRP3 inflammasome by Saikosaponin-d alleviates acute liver injury in carbon tetrachloride-induced hepatitis in mice. International Journal of Immunopathology and Pharmacology 34 2058738420950593. (https://doi.org/10.1177/2058738420950593)
Christgen S, Place DE & Kanneganti TD 2020 Toward targeting inflammasomes: insights into their regulation and activation. Cell Research 30 315–327. (https://doi.org/10.1038/s41422-020-0295-8)
Compan V, Baroja-Mazo A, López-Castejón G, Gomez AI, Martínez CM, Angosto D, Montero MT, Herranz AS, Bazán E & Reimers D et al.2012 Cell volume regulation modulates NLRP3 inflammasome activation. Immunity 37 487–500. (https://doi.org/10.1016/j.immuni.2012.06.013)
Cornut M, Bourdonnay E & Henry T 2020 Transcriptional regulation of inflammasomes. International Journal of Molecular Sciences 21 8087. (https://doi.org/10.3390/ijms21218087)
Daniels MJD, Rivers-Auty J, Schilling T, Spencer NG, Watremez W, Fasolino V, Booth SJ, White CS, Baldwin AG & Freeman S et al.2016 Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer’s disease in rodent models. Nature Communications 7 12504. (https://doi.org/10.1038/ncomms12504)
Danielski LG, Giustina AD, Bonfante S, Barichello T & Petronilho F 2020 The NLRP3 inflammasome and its role in sepsis development. Inflammation 43 24–31. (https://doi.org/10.1007/s10753-019-01124-9)
Di AX, Ye S, Malireddi Z, Kometani RKS, Zhong S, Mittal M, Hong M, Kanneganti Z, Rehman T-D & Malik J et al.2018 The TWIK2 potassium efflux channel in macrophages mediates NLRP3 inflammasome-induced inflammation. Immunity 49 56.e4–65.e4. (https://doi.org/10.1016/j.immuni.2018.04.032)
Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, Sun H, Wang DC & Shao F 2016 Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 535 111–116. (https://doi.org/10.1038/nature18590)
Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT & Tschopp J 2008 Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 320 674–677. (https://doi.org/10.1126/science.1156995)
Elliott EI & Sutterwala FS 2015 Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunological Reviews 265 35–52. (https://doi.org/10.1111/imr.12286)
Escames G, López LC, Ortiz F, López A, García JA, Ros E & Acuña-Castroviejo D 2007 Attenuation of cardiac mitochondrial dysfunction by melatonin in septic mice. FEBS Journal 274 2135–2147. (https://doi.org/10.1111/j.1742-4658.2007.05755.x)
Exline MC & Crouser ED 2008 Mitochondrial mechanisms of sepsis-induced organ failure. Frontiers in Bioscience 13 5030–5041. (https://doi.org/10.2741/3061)
Fan P, Xie XH, Chen CH, Peng X, Zhang P, Yang C & Wang YT 2019 Molecular regulation mechanisms and interactions between reactive oxygen species and mitophagy. DNA and Cell Biology 38 10–22. (https://doi.org/10.1089/dna.2018.4348)
Franchi L, Kanneganti TD, Dubyak GR & Núñez G 2007 Differential requirement of P2X7 receptor and intracellular K+ for caspase-1 activation induced by intracellular and extracellular bacteria. Journal of Biological Chemistry 282 18810–18818. (https://doi.org/10.1074/jbc.M610762200)
Franchi L, Eigenbrod T, Muñoz-Planillo R & Nuñez G 2009 The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nature Immunology 10 241–247. . (https://doi.org/10.1038/ni.1703)
Gao YL, Zhai JH & Chai YF 2018 Recent advances in the molecular mechanisms underlying pyroptosis in sepsis. Mediators of Inflammation 2018 5823823. (https://doi.org/10.1155/2018/5823823)
Gong T, Yang Y, Jin T, Jiang W & Zhou R 2018 Orchestration of NLRP3 inflammasome activation by ion fluxes. Trends in Immunology 39 393–406. (https://doi.org/10.1016/j.it.2018.01.009)
Groß CJ, Mishra R, Schneider KS, Médard G, Wettmarshausen J, Dittlein DC, Shi H, Gorka O, Koenig PA & Fromm S et al.2016 K+ efflux-independent NLRP3 inflammasome activation by small molecules targeting mitochondria. Immunity 45 761–773. (https://doi.org/10.1016/j.immuni.2016.08.010)
Gross O, Poeck H, Bscheider M, Dostert C, Hannesschläger N, Endres S, Hartmann G, Tardivel A, Schweighoffer E & Tybulewicz V et al.2009 Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature 459 433–436. (https://doi.org/10.1038/nature07965)
Gustafsson ÅB & Dorn 2nd GW 2019 Evolving and expanding the roles of mitophagy as a homeostatic and pathogenic process. Physiological Reviews 99 853–892. (https://doi.org/10.1152/physrev.00005.2018)
Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, Tardivel A, Heuschling P & Dostert C 2015 NLRP3 inflammasome is expressed and functional in mouse brain microglia but not in astrocytes. PLoS ONE 10 e0130624. (https://doi.org/10.1371/journal.pone.0130624)
Hafner-Bratkovič I & Pelegrín P 2018 Ion homeostasis and ion channels in NLRP3 inflammasome activation and regulation. Current Opinion in Immunology 52 8–17. (https://doi.org/10.1016/j.coi.2018.03.010)
Hamacher-Brady A & Brady NR 2016 Mitophagy programs: mechanisms and physiological implications of mitochondrial targeting by autophagy. Cellular and Molecular Life Sciences 73 775–795. (https://doi.org/10.1007/s00018-015-2087-8)
Hardy G, Hardy I & Manzanares W 2012 Selenium supplementation in the critically ill. Nutrition in Clinical Practice 27 21–33. (https://doi.org/10.1177/0884533611434116)
Harrington JS, Choi AMK & Nakahira K 2017 Mitochondrial DNA in sepsis. Current Opinion in Critical Care 23 284–290. (https://doi.org/10.1097/MCC.0000000000000427)
Harris J, Deen N, Zamani S & Hasnat MA 2018 Mitophagy and the release of inflammatory cytokines. Mitochondrion 41 2–8. (https://doi.org/10.1016/j.mito.2017.10.009)
Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA & Latz E 2008 Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nature Immunology 9 847–856. (https://doi.org/10.1038/ni.1631)
Hsu BG, Lee RP, Yang FL, Harn HJ & Chen HI 2006 Post-treatment with N-acetylcysteine ameliorates endotoxin shock-induced organ damage in conscious rats. Life Sciences 79 2010–2016. (https://doi.org/10.1016/j.lfs.2006.06.040)
Hu Q, Ren J, Wu J, Li G, Wu X, Liu S, Wang G, Gu G & Li J 2017 Elevated levels of plasma mitochondrial DNA are associated with clinical outcome in intra-abdominal infections caused by severe trauma. Surgical Infections 18 610–618. (https://doi.org/10.1089/sur.2016.276)
Huang TS, Shyu YC, Chen HY, Lin LM, Lo CY, Yuan SS & Chen PJ 2013 Effect of parenteral selenium supplementation in critically ill patients: a systematic review and meta-analysis. PLoS ONE 8 e54431. (https://doi.org/10.1371/journal.pone.0054431)
Jiménez-Sousa MA, Tamayo E, Guzmán-Fulgencio M, Heredia M, Fernández-Rodríguez A, Gómez E, Almansa R, Gómez-Herreras JI, García-Álvarez M & Gutiérrez-Junco S et al.2015 Mitochondrial DNA haplogroups are associated with severe sepsis and mortality in patients who underwent major surgery. Journal of Infection 70 20–29. (https://doi.org/10.1016/j.jinf.2014.07.005)
Jin HS, Suh HW, Kim SJ & Jo EK 2017 Mitochondrial control of innate immunity and inflammation. Immune Network 17 77–88. (https://doi.org/10.4110/in.2017.17.2.77)
Kalbitz M, Fattahi F, Grailer JJ, Jajou L, Malan EA, Zetoune FS, Huber-Lang M, Russell MW & Ward PA 2016 Complement-induced activation of the cardiac NLRP3 inflammasome in sepsis. FASEB Journal 30 3997–4006. (https://doi.org/10.1096/fj.201600728R)
Kang JW, Kim SJ, Cho HI & Lee SM 2015 DAMPs activating innate immune responses in sepsis. Ageing Research Reviews 24 54–65. (https://doi.org/10.1016/j.arr.2015.03.003)
Khaminets A, Behl C & Dikic I 2016 Ubiquitin-dependent and independent signals in selective autophagy. Trends in Cell Biology 26 6–16. (https://doi.org/10.1016/j.tcb.2015.08.010)
Khwaja B, Thankam FG & Agrawal DK 2021 Mitochondrial DAMPs and altered mitochondrial dynamics in OxLDL burden in atherosclerosis. Molecular and Cellular Biochemistry 476 1915–1928. (https://doi.org/10.1007/s11010-021-04061-0)
Kim JY & Lee SM 2004 Effect of ascorbic acid on hepatic vasoregulatory gene expression during polymicrobial sepsis. Life Sciences 75 2015–2026. (https://doi.org/10.1016/j.lfs.2004.06.002)
Kim JY & Lee SM 2006 Vitamins C and E protect hepatic cytochrome P450 dysfunction induced by polymicrobial sepsis. European Journal of Pharmacology 534 202–209. (https://doi.org/10.1016/j.ejphar.2006.01.015)
Kim MJ, Yoon JH & Ryu JH 2016 Mitophagy: a balance regulator of NLRP3 inflammasome activation. BMB Reports 49 529–535. (https://doi.org/10.5483/BMBRep.2016.49.10.115)
Kozlov AV & Grillari J 2022 Pathogenesis of multiple organ failure: the impact of systemic damage to plasma membranes. Frontiers in Medicine 9 806462. (https://doi.org/10.3389/fmed.2022.806462)
Kung CT, Hsiao SY, Tsai TC, Su CM, Chang WN, Huang CR, Wang HC, Lin WC, Chang HW & Lin YJ et al.2012 Plasma nuclear and mitochondrial DNA levels as predictors of outcome in severe sepsis patients in the emergency room. Journal of Translational Medicine 10 130. (https://doi.org/10.1186/1479-5876-10-130)
Lage SL, Amaral EP, Hilligan KL, Laidlaw E, Rupert A, Namasivayan S, Rocco J, Galindo F, Kellogg A & Kumar P et al.2021 Persistent oxidative stress and inflammasome activation in CD14highCD16- monocytes from COVID-19 patients. Frontiers in Immunology 12 799558. (https://doi.org/10.3389/fimmu.2021.799558)
Lamkanfi M & Dixit VM 2011 Modulation of inflammasome pathways by bacterial and viral pathogens. Journal of Immunology 187 597–602. (https://doi.org/10.4049/jimmunol.1100229)
Lamkanfi M & Dixit VM 2014 Mechanisms and functions of inflammasomes. Cell 157 1013–1022. (https://doi.org/10.1016/j.cell.2014.04.007)
Lang L, Xu B, Yuan J, Li S, Lian S, Chen Y, Guo J & Yang H 2020 GABA-mediated activated microglia induce neuroinflammation in the hippocampus of mice following cold exposure through the NLRP3 inflammasome and NF-κB signaling pathways. International Immunopharmacology 89 106908. (https://doi.org/10.1016/j.intimp.2020.106908)
Lazzaro A, De Girolamo G, Filippi V, Innocenti GP, Santinelli L, Ceccarelli G, Trecarichi EM, Torti C, Mastroianni CM & d’Ettorre G et al.2022 The interplay between host defense, infection, and clinical status in septic patients: a narrative review. International Journal of Molecular Sciences 23 803. (https://doi.org/10.3390/ijms23020803)
Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, Germain RN, Kastner DL & Chae JJ 2012 The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 492 123–127. (https://doi.org/10.1038/nature11588)
Li H, Willingham SB, Ting JP-Y & Re F 2008 Cutting edge: inflammasome activation by alum and alum’s adjuvant effect are mediated by NLRP3. Journal of Immunology 181 17–21. (https://doi.org/10.4049/jimmunol.181.1.17)
Lin L, Que R, Shen Y, Chen Y, Yan N & Li Y 2018 Saikosaponin-d alleviates carbon-tetrachloride induced acute hepatocellular injury by inhibiting oxidative stress and NLRP3 inflammasome activation in the HL-7702 cell line. Molecular Medicine Reports 17 7939–7946. (https://doi.org/10.3892/mmr.2018.8849)
Liu XH, Kwon D, Schielke GP, Yang GY, Silverstein FS & Barks JDE 1999 Mice deficient in interleukin-1 converting enzyme are resistant to neonatal hypoxic-ischemic brain damage. Journal of Cerebral Blood Flow and Metabolism 19 1099–1108. (https://doi.org/10.1097/00004647-199910000-00006)
Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H & Lieberman J 2016 Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 535 153–158. (https://doi.org/10.1038/nature18629)
Liu Q, Zhang D, Hu D, Zhou X & Zhou Y 2018 The role of mitochondria in NLRP3 inflammasome activation. Molecular Immunology 103 115–124. (https://doi.org/10.1016/j.molimm.2018.09.010)
Lopes-Pires ME, Frade-Guanaes JO & Quinlan GJ 2021 Clotting dysfunction in sepsis: a role for ROS and potential for therapeutic intervention. Antioxidants 11 88. (https://doi.org/10.3390/antiox11010088)
Man SM, Karki R & Kanneganti TD 2017 Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunological Reviews 277 61–75. (https://doi.org/10.1111/imr.12534)
Mantzarlis K, Tsolaki V & Zakynthinos E 2017 Role of oxidative stress and mitochondrial dysfunction in sepsis and potential therapies. Oxidative Medicine and Cellular Longevity 2017 5985209. (https://doi.org/10.1155/2017/5985209)
Manzanares W, Dhaliwal R, Jiang X, Murch L & Heyland DK 2012 Antioxidant micronutrients in the critically ill: a systematic review and meta-analysis. Critical Care 16 R66. (https://doi.org/10.1186/cc11316)
Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM & Dixit VM 2006 Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 440 228–232. (https://doi.org/10.1038/nature04515)
Martinon F, Pétrilli V, Mayor A, Tardivel A & Tschopp J 2006 Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440 237–241. (https://doi.org/10.1038/nature04516)
Misawa T, Takahama M, Kozaki T, Lee H, Zou J, Saitoh T & Akira S 2013 Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nature Immunology 14 454–460. (https://doi.org/10.1038/ni.2550)
Mishra V, Baines M, Perry SE, McLaughlin PJ, Carson J, Wenstone R & Shenkin A 2007 Effect of selenium supplementation on biochemical markers and outcome in critically ill patients. Clinical Nutrition 26 41–50. (https://doi.org/10.1016/j.clnu.2006.10.003)
Muñoz-Planillo R, Kuffa P, Martínez-Colón G, Smith BL, Rajendiran TM & Núñez G 2013 K⁺ efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity 38 1142–1153. (https://doi.org/10.1016/j.immuni.2013.05.016)
Murakami T, Ockinger J, Yu J, Byles V, McColl A, Hofer AM & Horng T 2012 Critical role for calcium mobilization in activation of the NLRP3 inflammasome. PNAS 109 11282–11287. (https://doi.org/10.1073/pnas.1117765109)
Nakamori Y, Park EJ & Shimaoka M 2020 Immune deregulation in sepsis and septic shock: reversing immune paralysis by targeting PD-1/PD-L1 pathway. Frontiers in Immunology 11 624279. (https://doi.org/10.3389/fimmu.2020.624279)
Ogilvie AC, Groeneveld ABJ, Straub JP & Thijs LG 1991 Plasma lipid peroxides and antioxidants in human septic shock. Intensive Care Medicine 17 40–44. (https://doi.org/10.1007/BF01708408)
Omer M, Melo AM, Kelly L, Mac Dermott EJ, Leahy TR, Killeen O, Saugstad OD, Savani RC & Molloy EJ 2020 Emerging role of the NLRP3 inflammasome and interleukin-1β in neonates. Neonatology 117 545–554. (https://doi.org/10.1159/000507584)
Orning P, Weng D, Starheim K, Ratner D, Best Z, Lee B, Brooks A, Xia S, Wu H & Kelliher MA et al.2018 Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science 362 1064–1069. (https://doi.org/10.1126/science.aau2818)
Osca-Verdegal R, Beltrán-García J, Pallardó FV & García-Giménez JL 2021 Role of microRNAs as biomarkers in sepsis-associated encephalopathy. Molecular Neurobiology 58 4682–4693. (https://doi.org/10.1007/s12035-021-02445-3)
Paik S, Kim JK, Silwal P, Sasakawa C & Jo EK 2021 An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cellular and Molecular Immunology 18 1141–1160. (https://doi.org/10.1038/s41423-021-00670-3)
Parihar A, Parihar MS, Milner S & Bhat S 2008 Oxidative stress and anti-oxidative mobilization in burn injury. Burns 34 6–17. (https://doi.org/10.1016/j.burns.2007.04.009)
Pétrilli V, Papin S, Dostert C, Mayor A, Martinon F & Tschopp J 2007 Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death and Differentiation 14 1583–1589. (https://doi.org/10.1038/sj.cdd.4402195)
Prauchner CA 2017 Oxidative stress in sepsis: pathophysiological implications justifying antioxidant co-therapy. Burns 43 471–485. (https://doi.org/10.1016/j.burns.2016.09.023)
Protter DSW & Parker R 2016 Principles and properties of stress granules. Trends in Cell Biology 26 668–679. (https://doi.org/10.1016/j.tcb.2016.05.004)
Rahmel T, Marko B, Nowak H, Bergmann L, Thon P, Rump K, Kreimendahl S, Rassow J, Peters J & Singer M et al.2020 Mitochondrial dysfunction in sepsis is associated with diminished intramitochondrial TFAM despite its increased cellular expression. Scientific Reports 10 21029. (https://doi.org/10.1038/s41598-020-78195-4)
Rossol M, Pierer M, Raulien N, Quandt D, Meusch U, Rothe K, Schubert K, Schöneberg T, Schaefer M & Krügel U et al.2012 Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein-coupled calcium sensing receptors. Nature Communications 3 1329. (https://doi.org/10.1038/ncomms2339)
Rottenberg H & Hoek JB 2017 The path from mitochondrial ROS to aging runs through the mitochondrial permeability transition pore. Aging Cell 16 943–955. (https://doi.org/10.1111/acel.12650)
Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N & Finfer S et al.2020 Global, regional, and national sepsis incidence and mortality, 1990–2017: analysis for the global burden of disease study. Lancet 395 200–211. (https://doi.org/10.1016/S0140-6736(1932989-7)
Rudyk O, Phinikaridou A, Prysyazhna O, Burgoyne JR, Botnar RM & Eaton P 2013 Protein kinase G oxidation is a major cause of injury during sepsis. PNAS 110 9909–9913. (https://doi.org/10.1073/pnas.1301026110)
Sakr Y, Reinhart K, Bloos F, Marx G, Russwurm S, Bauer M & Brunkhorst F 2007 Time course and relationship between plasma selenium concentrations, systemic inflammatory response, sepsis, and multiorgan failure. British Journal of Anaesthesia 98 775–784. (https://doi.org/10.1093/bja/aem091)
Samir P, Kesavardhana S, Patmore DM, Gingras S, Malireddi RKS, Karki R, Guy CS, Briard B, Place DE & Bhattacharya A et al.2019 DDX3X acts as a live-or-die checkpoint in stressed cells by regulating NLRP3 inflammasome. Nature 573 590–594. (https://doi.org/10.1038/s41586-019-1551-2)
Sarhan J, Liu BC, Muendlein HI, Li P, Nilson R, Tang AY, Rongvaux A, Bunnell SC, Shao F & Green DR et al.2018 Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. PNAS 115 E10888–E10897. (https://doi.org/10.1073/pnas.1809548115)
Sborgi L, Rühl S, Mulvihill E, Pipercevic J, Heilig R, Stahlberg H, Farady CJ, Müller DJ, Broz P & Hiller S 2016 GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. EMBO Journal 35 1766–1778. (https://doi.org/10.15252/embj.201694696)
Schneck E, Edinger F, Hecker M, Sommer N, Pak O, Weissmann N, Hecker A, Reichert M, Markmann M & Sander M et al.2020 Blood levels of free-circulating mitochondrial DNA in septic shock and postsurgical systemic inflammation and its influence on coagulation: a secondary analysis of a prospective observational study. Journal of Clinical Medicine 9 2056. (https://doi.org/10.3390/jcm9072056)
Schorn C, Frey B, Lauber K, Janko C, Strysio M, Keppeler H, Gaipl US, Voll RE, Springer E & Munoz LE et al.2011 Sodium overload and water influx activate the NALP3 inflammasome. Journal of Biological Chemistry 286 35–41. (https://doi.org/10.1074/jbc.M110.139048)
Sharma D & Kanneganti TD 2016 The cell biology of inflammasomes: mechanisms of inflammasome activation and regulation. Journal of Cell Biology 213 617–629. (https://doi.org/10.1083/jcb.201602089)
Sharma A, Tate M, Mathew G, Vince JE, Ritchie RH & de Haan JB 2018 Oxidative stress and NLRP3-Inflammasome activity as significant drivers of diabetic cardiovascular complications: therapeutic implications. Frontiers in Physiology 9 114. (https://doi.org/10.3389/fphys.2018.00114)
Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F & Shao F 2015 Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526 660–665. (https://doi.org/10.1038/nature15514)
Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L & Ojcius DM et al.2012 Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36 401–414. (https://doi.org/10.1016/j.immuni.2012.01.009)
Singer M 2014 The role of mitochondrial dysfunction in sepsis-induced multi-organ failure. Virulence 5 66–72. (https://doi.org/10.4161/viru.26907)
Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD & Coopersmith CM et al.2016 The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA 315 801–810. (https://doi.org/10.1001/jama.2016.0287)
Sok SPM, Ori D, Wada A, Okude H, Kawasaki T, Momota M, Nagoor NH & Kawai T 2021 1′-Acetoxychavicol acetate inhibits NLRP3-dependent inflammasome activation via mitochondrial ROS suppression. International Immunology 33 373–386. (https://doi.org/10.1093/intimm/dxab016)
Summersgill H, England H, Lopez-Castejon G, Lawrence CB, Luheshi NM, Pahle J, Mendes P & Brough D 2014 Zinc depletion regulates the processing and secretion of IL-1β. Cell Death and Disease 5 e1040. (https://doi.org/10.1038/cddis.2013.547)
Sutterwala FS, Haasken S & Cassel SL 2014 Mechanism of NLRP3 inflammasome activation. Annals of the New York Academy of Sciences 1319 82–95. (https://doi.org/10.1111/nyas.12458)
Takeda K, Shimada Y, Amano M, Sakai T, Okada T, Yoshiya I & Takeda IM 1984 Plasma lipid peroxides and alpha-tocopherol in critically ill patients. Critical Care Medicine 12 957–959. (doi:10.1097/00003246-198411000-00007.
Tang T, Lang X, Xu C, Wang X, Gong T, Yang Y, Cui J, Bai L, Wang J & Jiang W et al.2017 CLICs-dependent chloride efflux is an essential and proximal upstream event for NLRP3 inflammasome activation. Nature Communications 8 202. (https://doi.org/10.1038/s41467-017-00227-x)
Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR & Aunins J 1992 A novel heterodimeric cysteine protease is required for interleukin-1 β processing in monocytes. Nature 356 768–774. (https://doi.org/10.1038/356768a0)
Tschopp J 2011 Mitochondria: sovereign of inflammation? European Journal of Immunology 41 1196–1202. (https://doi.org/10.1002/eji.201141436)
Verhoef PA, Kertesy SB, Lundberg K, Kahlenberg JM & Dubyak GR 2005 Inhibitory effects of chloride on the activation of caspase-1, IL-1β secretion, and cytolysis by the P2X7 receptor. Journal of Immunology 175 7623–7634. (https://doi.org/10.4049/jimmunol.175.11.7623)
Wang X, Jiang W, Yan Y, Gong T, Han J, Tian Z & Zhou R 2014 RNA viruses promote activation of the NLRP3 inflammasome through a RIP1-RIP3-DRP1 signaling pathway. Nature Immunology 15 1126–1133. (https://doi.org/10.1038/ni.3015)
Wei H, Liu L & Chen Q 2015 Selective removal of mitochondria via mitophagy: distinct pathways for different mitochondrial stresses. Biochimica et Biophysica Acta: Molecular Cell Research 1853 2784–2790. (https://doi.org/10.1016/j.bbamcr.2015.03.013)
West AP & Shadel GS 2017 Mitochondrial DNA in innate immune responses and inflammatory pathology. Nature Reviews: Immunology 17 363–375. (https://doi.org/10.1038/nri.2017.21)
Wu X, Dong L, Lin X & Li J 2017 Relevance of the NLRP3 inflammasome in the pathogenesis of chronic liver disease. Frontiers in Immunology 8 1728. (https://doi.org/10.3389/fimmu.2017.01728)
Wu Y, Yao YM & Lu ZQ 2019 Mitochondrial quality control mechanisms as potential therapeutic targets in sepsis-induced multiple organ failure. Journal of Molecular Medicine 97 451–462. (https://doi.org/10.1007/s00109-019-01756-2)
Wu Y, Hao C, Liu X, Han G, Yin J, Zou Z, Zhou J & Xu C 2020 MitoQ protects against liver injury induced by severe burn plus delayed resuscitation by suppressing the mtDNA-NLRP3 axis. International Immunopharmacology 80 106189. (https://doi.org/10.1016/j.intimp.2020.106189)
Xian H, Liu Y, Rundberg Nilsson A, Gatchalian R, Crother TR, Tourtellotte WG, Zhang Y, Aleman-Muench GR, Lewis G & Chen W et al.2021 Metformin inhibition of mitochondrial ATP and DNA synthesis abrogates NLRP3 inflammasome activation and pulmonary inflammation. Immunity 54 1463–1477.e11. (https://doi.org/10.1016/j.immuni.2021.05.004)
Xu Y, Shen J & Ran Z 2020 Emerging views of mitophagy in immunity and autoimmune diseases. Autophagy 16 3–17. (https://doi.org/10.1080/15548627.2019.1603547)
Yang Y, Wang H, Kouadir M, Song H & Shi F 2019 Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death and Disease 10 128. (https://doi.org/10.1038/s41419-019-1413-8)
Yaron JR, Gangaraju S, Rao MY, Kong X, Zhang L, Su F, Tian Y, Glenn HL & Meldrum DR 2015 K(+) regulates Ca(2+) to drive inflammasome signaling: dynamic visualization of ion flux in live cells. Cell Death and Disease 6 e1954. (https://doi.org/10.1038/cddis.2015.277)
Yazdi AS, Guarda G, Riteau N, Drexler SK, Tardivel A, Couillin I & Tschopp J 2010 Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1α and IL-1β. PNAS 107 19449–19454. (https://doi.org/10.1073/pnas.1008155107)
Yu JW & Lee MS 2016 Mitochondria and the NLRP3 inflammasome: physiological and pathological relevance. Archives of Pharmacal Research 39 1503–1518. (https://doi.org/10.1007/s12272-016-0827-4)
Yue R, Zheng Z, Luo Y, Wang X, Lv M, Qin D, Tan Q, Zhang Y, Wang T & Hu H 2021 NLRP3-mediated pyroptosis aggravates pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction in mice: cardioprotective role of irisin. Cell Death Discovery 7 50. (https://doi.org/10.1038/s41420-021-00434-y)
Yuk JM, Silwal P & Jo EK 2020 Inflammasome and mitophagy connection in health and disease. International Journal of Molecular Sciences 21 4714. (https://doi.org/10.3390/ijms21134714)
Zhang YY & Ning BT 2021 Signaling pathways and intervention therapies in sepsis. Signal Transduction and Targeted Therapy 6 407. (https://doi.org/10.1038/s41392-021-00816-9)
Zhang JJ, Meng X, Li Y, Zhou Y, Xu DP, Li S & Li HB 2017 Effects of melatonin on liver injuries and diseases. International Journal of Molecular Sciences 18 673. (https://doi.org/10.3390/ijms18040673)
Zhang H, Feng YW & Yao YM 2018 Potential therapy strategy: targeting mitochondrial dysfunction in sepsis. Military Medical Research 5 41. (https://doi.org/10.1186/s40779-018-0187-0)
Zhang D, Yang XJ, Luo QD, Fu DL, Li ZL, Zhang P & Chong T 2019 Down-regulation of circular RNA_000926 attenuates renal cell carcinoma progression through miRNA-411-dependent CDH2 inhibition. American Journal of Pathology 189 2469–2486. (https://doi.org/10.1016/j.ajpath.2019.06.016)
Zhao Y, Huang S, Liu J, Wu X, Zhou S, Dai K & Kou Y 2018 Mitophagy contributes to the pathogenesis of inflammatory diseases. Inflammation 41 1590–1600. (https://doi.org/10.1007/s10753-018-0835-2)
Zhong Z, Zhai Y, Liang S, Mori Y, Han R, Sutterwala FS & Qiao L 2013 TRPM2 links oxidative stress to NLRP3 inflammasome activation. Nature Communications 4 1611. (https://doi.org/10.1038/ncomms2608)
Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin XJ, Wong J, Ding S, Seki E & Schnabl B et al.2018 New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560 198–203. (https://doi.org/10.1038/s41586-018-0372-z)