Open Access

Focus on the role of mitochondria in NLRP3 inflammasome activation: A prospective target for the treatment of ischemic stroke (Review)

  • Authors:
    • Xiaolu Zhang
    • Wenyun Zeng
    • Yue Zhang
    • Qun Yu
    • Miao Zeng
    • Jiali Gan
    • Wenlan Zhang
    • Xijuan Jiang
    • Huhu Li
  • View Affiliations

  • Published online on: April 7, 2022     https://doi.org/10.3892/ijmm.2022.5130
  • Article Number: 74
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Post‑ischemic neuroinflammation induced by the innate local immune response is a major pathophysiological feature of cerebral ischemic stroke, which remains the leading cause of mortality and disability worldwide. NLR family pyrin domain containing (NLRP)3 inflammasome crucially mediates post‑ischemic inflammatory responses via its priming, activation and interleukin‑1β release during hypoxic‑ischemic brain damage. Mitochondrial dysfunctions are among the main hallmarks of several brain diseases, including ischemic stroke. In the present review, focus was addressed on the role of mitochondria in cerebral ischemic stroke while keeping NLRP3 inflammasome as a link. Under ischemia and hypoxia, mitochondria are capable of controlling NLRP3 inflammasome‑mediated neuroinflammation through mitochondrial released contents, mitochondrial localization and mitochondrial related proteins. Thus, inflammasome and mitochondria may be attractive targets to treat ischemic stroke as well as the several drugs that target the process of mitochondrial function to treat cerebral ischemic stroke. At present, certain drugs have already been studied in clinical trials.

1. Introduction

Ischemic stroke is a common cause of disability, normally manifested as long-term neurological impairment, and even death (1,2). The pathogenesis of ischemic stroke is mainly caused by atherothrombosis at large cervical or intracranial arteries or by occlusion of cardio-embolus (3), which results in insufficient oxygen and glucose delivery to meet the requirement of cellular respiration (4). To date, the approved therapies of ischemic stroke are intravenous thrombolysis or thrombectomy (5), which can only be applied to a very small fraction of patients due to the narrow time window and strict indication criteria (6). As such, there is an urgent need to develop novel and alternative treatment strategies to treat ischemic stroke.

Post-ischemic neuroinflammation is an important pathological hallmark that affects both the development and prognosis of ischemic stroke (7,8). Once the inflammatory cascade is turned on, it aggravates neuron dysfunction and induces breakdown of blood brain barrier, brain edema and ultimately neuron death (9). Inflammasomes are intracellular multiprotein complexes that drive the activation of inflammatory responses. Among all types of inflammasomes, such as NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLR family CARD domain-containing protein 4 (NLRC4), and absent in melanoma 2 (AIM2), NLRP3 is the most studied one, particularly in the central neural system (10). It was shown that NLRP3 inflammasome activation regulates inflammatory response and accelerates neuron damage (11), and it acts as a key intermediate of neuroinflammation during the progress of ischemic stroke (12). Thus, inhibition of NLRP3 inflammasome activation may be applied as a novel treatment strategy for ischemic stroke (13).

Alternatively, mitochondrion is an organelle that plays roles in energy conversion and metabolism, and its dysfunction is the crucial pathophysiological change in ischemic stroke due to oxygen-glucose deprivation (14). It was found that a few studies have confirmed that NLRC4 (15), NLRP1 (16) and AIM2 (17) can promote cerebral ischemic injury, but there is no evidence that mitochondria can affect the pathological process of cerebral ischemia through them. Previous studies revealed that mitochondrial dysfunction is a vital event during the NLPR3 inflammasome activation (18-20). However, the role of NLRP3 inflammasome in sensing mitochondrial damage and how mitochondria trigger NLRP3 activation in ischemic stroke is to be elucidated (21). The present review focused on the role of mitochondria in NLRP3 inflammasome activation under ischemic stroke and described the currently used drugs and potential treatment targets for ischemic stroke.

2. Pathophysiological changes of mitochondria in ischemic stroke

The most sensitive organ suffering from ischemia and hypoxia is the brain (22), which depends on continuous delivery of oxygen and nutrients to maintain its function (23). Adult brain accounts for 20% of all the oxygen consumed by the whole body, with merely 2% of the total body weight (24). Given the high-energy demanding nature of neurons in the brain, it is essential that the mitochondrial pool remains healthy and provides a continuous and efficient supply of energy (25). Mitochondria produce most ATP production through mitochondrial respiratory chain (MRC) and oxidative phosphorylation (OXPHOS). However, mitochondria are very sensitive to ischemia and hypoxia (26); under these circumstances, they undergo significant alterations, including Ca2+ influx, mitochondrial permeability transition pore (mPTP) opening, reactive oxygen species (ROS) generation, DNA damage and mutation, unbalanced mitochondrial dynamics, and aberrant mitochondrial position (27-29). These changes in mitochondria are termed as mitochondrial dysfunction, which is strongly implicated in patients and multiple animal models with ischemic stroke (30-34).

During ischemic stroke, ischemia and hypoxia reduce ATP production and lead to dysfunction of the Na+/K+ ATPase pump (35). Within min following the onset of cerebral ischemia, ATP depletion deactivates the sodium-potassium pump and then excessive glutamate is released into extracellular fluid (36). Glutamate, as the main excitatory neurotransmitter in the central nervous system, is essential to multiple functions of neurons by binding to different types of receptors including N-methyl-d-aspartate receptor and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (37). Under ischemia-hypoxia condition, the high level of extracellular glutamate induces massive calcium influx, which further aggravates mitochondrial calcium overload (38). Elevated mitochondrial calcium launches a series of events ranging from mPTP opening and dissipation of ΔΨm to excessive ROS production, leading to neuroinflammation and eventually neuronal death (Fig. 1) (28). While Inhibition of the heat shock 75-kDa glucose-regulated protein was able to effectively ameliorate mitochondrial calcium overload and alleviate the ischemic stroke (39).

Figure 1

Mitochondria play a pivotal role in the pathological process upon ischemic insult. ATP depletion due to OGD leads to Na+/K+ ATPase pump dysfunction. This causes the depolarization of neuronal membrane to release excessive glutamate. In turn, glutamate receptors, such as NMDA and AMPA, are over activated, resulting in calcium influx into neurons. TRPM2 channel, which is a glutamate-independent ion channel, also leads to intracellular Ca2+ calcium overload which induces mtROS release. Finally, ROS release and mitochondrial dysfunction ensures to initiate inflammation. The production of ROS leads to the dissociation of TXNIP from TRX to active NLRP3 inflammation. RIPK1 interacts with MCU to upregulate mitochondrial Ca2+ uptake and disrupts the mitochondrial membrane integrity. Furthermore, ischemia triggers the depolarization of ∆Ψm and induction of mPTP, which leads to the production of mitochondrial DAMPs (such as cardiolipin and mtDNA). Under the stimulation of risk factors, such as lipopolysaccharide and nigericin, SHP2 enters cells and binds to ANT1, thus stabilizing mPTPs and inhibiting the release of mtROS and mtDNA. The dissociation of Nrf2 from Keap1 can inhibit mtROS release. These can cause the activation of NLRP3 inflammasome and contribute to tissue damage following ischemic stroke injury. OGD, oxygen-glucose deprivation; NMDA, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate; AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate; mtROS, mitochondrial ROS; ROS, reactive oxygen species; TXNIP, thioredoxin interacting protein; TRX, thioredoxin; NLRP3, NLR family pyrin domain containing 3; RIPK1, necrosome-containing receptor-interacting protein kinase 1; MCU, mitochondrial Ca2+ uniporter; mPTP, mitochondrial permeability transition pore; DAMP, damage-associated molecular pattern; SHP2, Src homology 2 domain-containing tyrosine phosphatase-2; ANT1, adenine nucleotide transferase 1; mtDNA mitochondrial DNA.

Meanwhile, the decreased ATP depletes nicotinamide adenine dinucleotide (NAD+), whose bioenergy state is a critical determinant for neuronal survival under excitotoxicity and ischemia (40). The reduced NAD+ motivates mitochondria to the vicinity of endoplasmic reticulum (ER) to form mitochondria-associated ER membranes (MAMs) (41). The diverse drugs or compounds with clear anti-stroke effects were revealed to reduce or partially reverse neuronal damage through inhibition of MAM-related proteins following ischemic stroke (42,43). Moreover, certain MAM-related proteins join mPTP to regulate its opening (44), which is an important marker of cerebral cell death during ischemia/reperfusion. Once ischemia/reperfusion injury promotes mPTP opening, mitochondrial ROS (mtROS), mitochondrial DNA (mtDNA) and cardiolipin inside the mitochondria are released into the cytoplasm, where they act as danger signals when recognized by innate immune receptors to exacerbate ischemia damage (45).

Besides energy production, mitochondria also generate a small amount of ROS, which induces oxidative damage (46). Increased ROS was generated during cerebral ischemia, particularly during reperfusion by disrupting mitochondrial electron transport (47,48). The broken mitochondria produce large amounts of ROS, which in turn affect the function of adjacent mitochondria. Since mitochondria are both the sources of ROS production and the targets of ROS, oxidant-induced mitochondrial dysfunction forms a 'vicious cycle' in patients with ischemic stroke (49,50). Furthermore, the ROS release results in oxidation and mutation of mtDNA, release of mitochondrial proteins and impaired mitophagy as shown in rat model of middle cerebral artery occlusion (MCAO). Moreover, the accumulation of ROS promotes neuroinflammation and neuron apoptosis after oxygen-glucose deprivation/reoxygenation (OGD/R) (51).

In addition to mitochondrial function, changes in mitochondrial structure play an important role in the progress of ischemic stroke. Mitochondria are highly dynamic organelles to regulate their size, form and mtDNA integrity via continuous fission, fusion and mitophagy (52). When neurons undergo ischemia, mitochondrial fission and fusion are transitory to maintain its structure integrity and function. However, under the circumstance of large amount of ROS production (53) and mtDNA damage (54), mitochondria are divided excessively, resulting in mitochondrial dysfunction (55). Thus, mitochondrial fission occurs as an upstream and early event in brain cell death following ischemia (56,57).

Furthermore, mutation mtDNA genes is correlated mitochondrial dysfunction with genomic instability. mtDNA is markedly more prone to mutations than nuclear DNA (58). Particularly, the frequency of mtDNA mutations was found to be significantly higher in the brain of patients with ischemic stroke, and numerous of these mutations resulted in an alteration of amino acid therefore structure of protein (34). The vulnerability of mtDNA to mutation is due to its lacking of high-fidelity repair processes, the high-copy-number of mtDNA within each cell, and the close proximity to ROS-generating machinery (59). In a word, mutation of gene-encoded subunits in mtDNA results in more ROS generation, which turns mtDNA more susceptible to mutation.

3. NLRP3 inflammasome-mediated neuroinflammation in ischemic stroke

Neuroinflammation in ischemic stroke

Upon stroke attack, the interruption and reperfusion of blood flow in the brain tissue trigger the infiltration of inflammatory cells to induce neuronal death (60). Neuroinflammation is a primary pathological event involved in the process of ischemic injury and repair (61). In response to injury in the brain, neuroinflammation occurs in various cells, including neurons, microglia and astrocytes (62). In the acute phase, the damaged neurons and resident immune cells secrete inflammatory mediators and activate microglia, which are the first line of defense in the nervous system (63) that produce more inflammatory factors (62,64). Additionally, astrocytes that are activated by ischemia mediate inflammatory response to aggravate ischemic injury. However, they also limit the spread of inflammation by inhibiting excitotoxicity and secreting neurotrophic factors (65,66). Interestingly, NLRP3 inflammasome was found to play a key role in driving neuroinflammation in these cells during acute ischemic stroke, and early blockade of NLRP3 protects neurons from ischemia-reperfusion injury by mitigating inflammation (67).

The composition and activation of the NLRP3 inflammasome

NLRP3 inflammasome is the most widely described inflammatory complex, which is composed of NLRP3 as its receptor, apoptosis-associated speck-like protein containing a CARD (ASC) as its adaptor, and caspase-1 as its effector. Activation of NLRP3 inflammasome includes two steps, namely, priming and activation (68). The priming step is the recognition of pathogen-associated molecular pattern (PAMP) or damage-associated molecular pattern (DAMP) via pattern recognition receptors (PRRs). Transcription and post-translational modification (PTM) of NLRP3 inflammasome promote the transcription of NLRP3 and the precursor of caspase-1, interleukin (IL)-1β and IL-18 (69). The priming of NLRP3 inflammasome not only provides material for NLRP3 inflammasome activation, but also allows NLRP3 and ASC to form the inflammasome assembly (70) or to protect them from degradation (71) through various PTM including ubiquitylation, deubiquitylation and phosphorylation. Then, the activation step is required to initiate the NLRP3 inflammasome assembly and subsequent activation. Generally, NLRP3 is oligomerized via its NACHT domains once activated (72). NLRP3 trimerization rather than dimerization is necessary for the inflammasome activation (73). Subsequently, activated NLRP3 interacts and recruits the adaptor molecule ASC via PYD-PYD interaction (74). The polymerized ASC further recruits the enzyme caspase-1 through CARD-CARD interactions to initiate autocatalytic cleavage of caspase-1 (75). Notably, ASC oligomerization is a key step in caspase-1 activation and caspase-1-dependent pyrophosphorylation upon NLRP3 stimulation (76). Once activated, caspase-1 cleaves the inactive pro-IL-1β and pro-IL-18 to release their active form of cytokines IL-1β and IL-18 to induce neuroinflammation and pyroptosis (77).

NLRP3 inflammasome in ischemic stroke

NLRP3 inflammasome is generally expressed in immune organs and cells, and is also expressed in the central nervous system (78,79). Liu et al (80) found that NLRP3 was activated in the microglia of ischemia-reperfusion injury rat model (81). Moreover, recent study suggested that NLRP3 was also expressed in the endothelial cells, neurons, and astrocytes of ischemic brain (82). It was demonstrated that NLRP3 inflammasome was firstly activated in microglia and then expressed in microvascular endothelial cells and neurons of transient MCAO (tMCAO) rat model (83).

Elevated levels of enlarged infarct size, neurological function and brain infarct volume were observed in MCAO rats with activation of NLRP3 inflammasome compared with sham rats (84). Furthermore, IL-1β and IL-18 levels were increased in the ipsilateral brain of ischemia-reperfusion mice model as well as in the postmortem ipsilateral brain of patients with stroke (79). The NLRP3 inhibitor MCC95 significantly reduced the infarct volume in a dose-dependent manner, the expression of different pro-inflammatory cytokines and NLRP3 inflammasome components, indicating its neuroprotective effect in the MCAO mice (85). Furthermore, elevated expression of NLRP3, caspase-1, ASC and IL-1β were present in a murine model of cerebral ischemia, while caspase-1 inhibition by VX765 prevented these changes and neuronal death. In summary, NLRP3 inflammasome exerts essential functions in the pathogenesis of ischemic stroke.

4. Mitochondria regulate NLRP3 inflammasome activation during ischemic stroke

Mitochondria regulate NLRP3 inflammasome activation under cerebral ischemia in bidirectional mode. Mitochondria are involved in the initiation and activation of NLRP3 inflammasome, leading to neuroinflammation and pyroptotic cell death (86). On one hand, the opening of mPTPs in damaged mitochondria release DAMPs, such as ATP, mtROS, cardiolipin and mtDNA, which are common causes for NLRP3 inflammasome activation (87). The localization of mitochondrial and its membrane-associated proteins also takes positive participation in activation of NLRP3 inflammasome during cerebral ischemic damage. On the other hand, active mitophagy and fractional mitochondrial-related proteins such as Src homology 2 domain-containing tyrosine phosphatase 2 (SHP2), mitofusin 2 (Mfn2) and nuclear factor E2-related factor-2 (Nrf2) negatively regulate the NLRP3 inflammasome expression to protect brain from ischemic injury (88) (Fig. 1).

The positive role of mitochondria in NLRP3 inflamma-some activation. Mitochondrial-released contents. mtROS

Dysfunctional mitochondria produce large amounts of mtROS by transferring electrons from the MRC to molecular oxygen to form mtROS (77). MtROS are important in NLRP3 priming and activation. It was revealed that mtROS regulates NLRP3 initiation earlier than activation by upregulating its transcription (89). As a non-transcriptional priming signal, deubiquitination of NLRP3 depends on mtROS generation (90). Elimination of mtROS inhibits NLRP3 deubiquitination, in response to lipopolysaccharide stimulation (91,92). By contrast, mtROS induces NLRP3-dependent lysosomal damage and inflammasome activation, and promotes macrophages pyroptosis by inducing Gasdermin D oxidation (93), which can be reversed by scavenging mtROS (94).

Next, high concentrations of mtROS results in NLRP3 activation and IL-1β production (92). Furthermore, increased mtROS induced by ischemia-reperfusion injury leads to release IL-1β, IL-18 and caspase-1 by cleaving their precursors (95). Conversely, inhibiting mtROS formation or eliminating mtROS by antioxidants strongly impairs the activation of NLRP3 inflammasome and IL-1β release (96,97). It was reported that mtROS-NLRP3 signaling is activated in BV2 cells after OGD/R for 24 h. Inhibition of mtROS release suppresses NLRP3 activation and alleviates NLRP3-mediating damage in microglia of ischemia-reperfusion rats (33).

Cardiolipin

Apart from mtROS, the opening of mPTPs releases other mitochondria-related DAMPs like mtDNA and mitochondrial lipids during ischemic stroke (98). Among the different mitochondrial lipids, cardiolipin is an anionic phospholipid that localizes at the inner mitochondrial membrane and facilitates OXPHOS in mitochondria (99). Cardiolipin oxidation and hydrolysis are a key mechanism of ischemia-reperfusion-induced brain injury (100). Nowadays, cardiolipin is reported to be effective for triggering the activation of NLRP3 inflammasome (101) after acute ischemia (102). On one hand, cardiolipin directly interacts with both the N-terminal leucine-rich repeat (LRR) of NLRP3 and full-length of caspase-1 of NLRP3 inflammasome (86,103). On the other hand, NLRP3 is tethered to mitochondria by cardiolipin in an mTROs-dependent manner and is thereby activated (104). Either interference with cardiolipin synthesis or knockdown of cardiolipin specifically inhibits NLRP3 inflammasome activation (78).

mtDNA

mtDNA was recognized as one of the endogenous DAMPs, which is released from damaged mitochondria into the cytoplasm to activate NLRP3 inflammasome and induce pyroptosis. It was shown that mtDNA was indispensable for NLRP3 activation by mtROS after ischemia-reperfusion (105,106). In response to various NLRP3 activators, mtDNA is rapidly released into cytoplasm to be oxidized to oxidized mtDNA (ox-mtDNA). Then, ox-mtDNA specifically localizes to NLRP3 (106), and directly binds to NLRP3 to trigger NLRP3 inflammasome activation (107). It was identified that NLRP3 inflammasome is over-activated in aged individuals, due to increased production of mtROS and/or mtDNA (108). High level of mtDNA induces the interaction of ASC with NLRP3 and pro-caspase-1 and promotes neuronal pyroptosis in the hippocampus of rats with incomplete ischemia-reperfusion injury (109). Correspondingly, repairing mtDNA oxidative damage inhibits NLRP3 activation and reduces reperfusion-associated ischemic brain injury (110). These results imply that mtDNA interacts with NLRP3 inflammasome to form positive feedback during the development of ischemic stroke.

Mitochondrial localization. MAMs

ER-mitochondrial contact is mediated by a specific membrane structure, known as MAM, which plays key roles in material transfer and signal transduction, including Ca2+ signaling (111,112). Notably, increased MAM aggravates mitochondrial dysfunction and enhances ROS production (113). In addition, MAM has overarching roles in innate immune system. When NLRP3 localizes at ER membrane, it is in the resting state. By contrast, when it relocates to MAM, it switches to activated state and functions in detecting ROS production from damaged mitochondria (114). Recruitment of NLRP3 to mitochondria enhances the ability of mtROS to activate NLRP3 inflammasome. ASC predominantly localized to the mitochondria, is transferred to MAM under the stimulation of NLRP3 activators. Furthermore, colocalization of ASC with MAM requires the presence of NLRP3 and is Ca2+ dependent (115). Notably, mitochondrial damage by NLRP3 inflammasome activators leads to the accumulation of NLRP3 and ASC in MAM (116). Ischemia-reperfusion injury increases the expression of MAM-related proteins, which accelerated signal communication with mitochondria through MAM in the male C57BL/6 mice (117). Silencing MAM-related protein p66Shc protects the integrity of blood-brain barrier, reduces infarct area, relieves the neurological deficit, and improves the survival rate of mice after MCAO (118). In a word, MAM is an ideal site for NLRP3 inflammasome activation and assembly which accelerates the development of ischemic stroke (119).

Microtubules

Generally, the movement of mitochondria in neurons is considered to be mediated by microtubules or microfilaments. Microtubules, which are formed by polymerization of α and β tubulins, are cell cytoskeleton to support intracellular transport between various organelles, particularly those involved in mitochondrial transport (120). Microtubules undergo various PTMs to take part in the transportation between mitochondria and ER, as well as in the subcellular localization of NLRP3 and ASC. Correspondingly, NLRP3 inflammasome activators induce mitochondrial transport to ER through the microtubule system, thereby facilitating the transfer of ASC from mitochondria to ER to combine with NLRP3 (121). A previous study found that the acetylation of microtubule α-tubulin is a necessary step for NLRP3 inflammasome activation (41) (Fig. 2). Moreover, NAD+ is an endogenous small molecule that regulates the microtubules (122). NAD+ caused by reduced ATP production moves mitochondria through microtubules, therefore promotes the assembly of NLRP3 inflammasome (41). Nicotinamide partially increases cellular NAD+ levels and effectively protects neurons from ischemic damage (40).

Mitochondrial-related proteins
Thioredoxin interacting protein (TXNIP)

TXNIP is an endogenous inhibitor of the thioredoxin (TRX) system and is expressed in nearly all cytoplasm and mitochondria (123). MtROS was revealed to promote the combination of NLRP3 with TXNIP which is a critical step that links oxidative stress to neuroinflammation (124,125). In response to mtROS release, TXNIP dissociates from TRX and translocates to MAM, to bind with NLRP3 and induces NLRP3 inflammasome activation (126). Inhibition of TRX expression interrupts the interaction between TRX and TXNIP, thus promoting the binding of TXNIP to NLRP3 and triggers the assembly and oligomerization of the NLRP3 inflammasome (127). In addition, TXNIP expression is upregulated in patients with stroke and rat model of cerebral ischemia (123,128). The inactivation of TXNIP relieves neurological deficits, cerebral infarction and edema from ischemic damage (123). Finally, TRX inhibitor downregulates the expression of TXNIP and suppresses NLRP3 inflammasome activation in astrocytes with OGD/R (80).

Ca2+ and its receptors

Ca2+ concentration increases either by Ca2+ influx from the extracellular space as triggered by neuronal activity or by Ca2+ outflux from the ER (129). Ca2+ accumulation is the determining factor of mPTP opening, which is an important marker of nerve cell death during ischemia, ensues by release of mitochondrial components that activate NLRP3 inflammasome (130). It has been revealed that NLRP3 receptors are activated by increased intracellular Ca2+ concentration in vitro and in vivo (131). Furthermore, when Ca2+ concentration in the cytoplasm increases, mitochondria uptake a large amount of Ca2+ through calcium-sensitive receptor (CASR) to activate the NLRP3 inflammasome. Consistently, knockdown of CASR reduces inflammasome activation. Moreover, a CASR agonist upregulates the expression of NLRP3, cleaved caspase-1, and IL-1β in the ipsilateral cortex of mice after stroke (132). Intriguingly, high intracellular Ca2+ concentration can also lead to Ca2+ influx to mitochondria through mitochondrial calcium uniporter (MCU), then loss of ΔΨm, resulting in NLRP3 inflammasome activation and IL-1β secretion (133). Murakami et al (134) proposed that Ca2+ signaling is an inter-mediate step of NLRP3 inflammasome activation. Similarly, transient receptor potential melastain-2 (TRPM2) channel is a glutamate-independent ion channel. Under the condition of ischemia injury, TRPM2 channel can be activated and mediate the transport of Ca2+, leading to intracellular calcium overload. Notably, intracellular calcium level further drives TRPM2 activity. In addition, TRPM2 promotes NLRP3 activation in OGD-induced neuronal injury, which was abolished by TRPM2 knockdown (135).

Necrosome-containing receptor-interacting protein kinase 1 (RIPK1)

RIPK1 is considered an essential regulator of apoptosis, necroptosis and inflammatory response. Under cerebral ischemia-hypoxia condition, RIPK1 induces necrotic apoptosis and neuroinflammation by destroying plasma membrane of endotheliocyte and microglia (136). Subsequently, released DAMPs from brain cells cause secondary inflammatory response, which aggravates ischemic damage (137,138). Upon being transported to out membrane of mitochondria, RIPK1 interacts with MCU to upregulate mitochondrial Ca2+ uptake, which leads to mtROS generation and NLRP3 activation (139). RIPK1 is upregulated in rat brain upon MCAO and localized to the microglia. Furthermore, RIPK1 triggers activation of NLRP3 inflammasome by disrupting the mitochondrial membrane integrity and by promoting mtROS release in ischemic microglia (140). As the first selective inhibitor, necrostatin-1 (Nec-1) significantly decreased RIPK1 phosphorylation in rat brain following ischemic stroke. Consistently, Nec-1 hindered IL-1β maturation in ischemic brains of rats (141).

Dynamin-related protein 1 (Drp1)

Mitochondrial dynamics are regulated by specific proteins through mitochondrial fission and fusion (142) which play an important role in NLRP3 inflammasome assembly and activation (143). Mitochondrial fission is primarily contributed to Drp1 activation (144), which promotes mitochondrial Ca2+ uptake (145) to trigger NLRP3 inflammasome activation. Consistently, inhibition of Drp1-dependent mitochondrial fission protects neurons from ischemic stroke by preserving the activity of respiratory chain, reducing superoxide production and delaying Ca2+ dysregulation (146). Moreover, inhibition of Drp1 withholds NLRP3 inflammasome activation and protects mitochondrial integrity to exert its neuroprotective effects (147). A recent study showed that neuronal death was prevented in MCAO rat model by lowering Drp1 expression and NLRP3 signal transduction (148).

The negative role of mitochondria in NLRP3 inflammasome activation
Mitophagy

Following ischemia-reperfusion injury, the damaged mitochondria are removed by autophagy-related mechanism, which is known as mitophagy (149). Mitophagy is an important mechanism of mitochondrial renewal and metabolism which downregulates the number and controls quality of mitochondria, induces mitochondria to maintain dynamic homeostasis (150) and consequently reduces mitochondria-dependent neuronal cell death (151). In previous studies, it was identified that induction of mitophagy protects against cerebral ischemia-reperfusion injury by inhibiting NLRP3 inflammasome activation (149,152). Following ischemia-reperfusion, mitophagy increased locally reduces the neuroinflammatory response induced by NLRP3 inflammasome to relieve neurological deficits (153). Blockage of mitophagy enhances the activation of the NLRP3 inflammasome (154). The damaged and dysfunctional mitochondria enhance NLRP3 inflammasome activation upon treatment with mitophagy inhibitors (116).

Mitochondrial transfer

Astrocytes (AS) are the most abundant glial cells in the central nervous system (155). When ischemic stroke occurs, AS provide energy support for injured neurons through the changes of its own bioenergy and mitochondrial dynamics (156). In addition, AS sense stress, transfer mitochondria as a 'help me' signal to adjacent injured neurons (157) and rescue damaged neurons from mitochondrial dysfunction to deal with stress (158). Concurrently, neurons also release damaged mitochondria and transfer them to as for endocytosis and degradation (159), so as to realize the recycling of mitochondria, making mitochondria crosstalk between healthy cells and damaged cells.

SHP2

SHP2 is a negative regulator of NLRP3 inflammasome activation (160). In response to NLRP3 inflammasome stimuli, SHP2 translocates to mitochondria and binds to adenine nucleotide transferase 1 (ANT1), which prevents the opening of mPTPs and the subsequent release of mtDNA and mtROS, thereby inhibiting NLRP3 inflammasome over-activation (160). In a focal cerebral ischemia model, ischemia-induced neuronal damage and death were significantly increased in nestin-SHP2-CS mice (SHP2 function was selectively removed from central nervous system) compared with wild-type mice. Additionally, transgenic mice that overexpress SHP2 are more sensitive to ischemia-induced brain damage (161).

Mfn2

Mfn2, a mitochondrial outer membrane protein, plays a pivotal part in mitochondrial fusion. It was reported that Mfn2 is downregulated while NLRP3 inflammasome and pyroptosis are activated in microglia and astrocytes of rats upon ischemia-reperfusion injury (162). Mfn2 overexpression attenuates free fatty acids induced mitochondrial damage, decreases mtROS production and inhibits NLRP3 inflammasome activation (163). Additionally, Mfn2 improves hypoxia induced neuronal apoptosis and prolongs the treatment time window of ischemic stroke by mitochondrial pathway (164).

Nrf2

Nrf2 is a well-known transcription factor that dissociates from Keap1 then translocates into the nucleus to initiate gene transcription via binding to antioxidant response element (ARE) during cellular stress conditions (165,166). The knockout of Nrf2 aggravates oxidative stress and inflammation (167). Nrf2 abates NLRP3 inflammasome activation by inhibiting the priming step to limit the assembly of NLRP3 inflammasome (96,168). Following cerebral ischemia-reperfusion, the activated Nrf2/ARE pathway inhibits ROS-induced NLRP3 inflammasome activation in BV2 microglia (169). Nrf2 knockout mice showed larger infarct size following ischemia-reperfusion, compared with that of control counterparts (170). Moreover, Nrf2 siRNA increased expressions of TXNIP, NLRP3, caspase-1 and IL-1β in brain of MCAO rats (171). Consequently, Nrf2 protects against NLRP3 inflammasome activation by regulating the TRX/TXNIP complex during cerebral ischemia-reperfusion injury (172).

5. Drugs that target mitochondria

Tissue plasminogen activator (tPA) and tPA recombinant protein were both used to treat ischemic stroke. However, these treatments have dangerous complications regarding reperfusion-injury (173). Reperfusion-injury generally causes oxidative stress, calcium overload and excitatory toxicity (174). At present, edaravone, butylphenol and other drugs are often used in the clinical treatment of stroke to alleviate the brain injury caused by calcium overload and excess ROS. In addition, mitochondrial dysfunction is deemed as a marker of ischemic stroke (175). Therefore, drugs that target mitochondria and directly or indirectly affect NLRP3 inflammasome to alleviate brain injury from the aspects of ROS and calcium overload are summarized in the present review (Table I).

Table I

Drugs that target mitochondria and NLRP3 inflammasome.

Table I

Drugs that target mitochondria and NLRP3 inflammasome.

DrugMitochondrial associated protein targetsNLRP3 inflammasome targetsModel(Refs.)
UmbelliferoneROS/TXNIP NLRP3/IL-1β/Caspase-1/IL-18MCAO/R(176)
CurcuminROS/TXNIP NLRP3/IL-1β/Caspase-1MCAO(177)
RuscogeninROS/TXNIP NLRP3/IL-1β/Caspase-1MCAO/R OGD/R(178)
resveratrolTXNIP NLRP3/IL-1β/Caspase-1eMCAO(179)
tPATXNIP NLRP3/IL-1β/ASC/Caspase-1t-MCAO(180)
Z-GuggulsteroneTXNIP NLRP3/IL-1β/IL-18MCAO/OGD(181)
Malibatol AROSIL-1βMCAO(182)
SesamolROSIL-1βMCAO/R(183)
MinocyclineROS NLRP3/IL-1β/IL-18tMCAO/R OGD/R(184)
SinomenineROS NLRP3/ASC/IL-1β/Caspase-1/IL-18MCAO/OGD(185)
IrisinROS NLRP3/IL-1β/Caspase-1OGD(186)
apocyninROS NLRP3/IL-18/IL-1β/ASC/Caspase-1MCAO/R(187)
RosuvastatinROSNLRP3MCAO(188)
arginaseROSIL-1βMCAO(189)
MedioresinolmtROS IL-1β/NLRP3/Caspase-1/ASC/tMCAO(190)
miR-668Drp-1/ROSNLRP3/IL-1βtMCAO/R OGD/R(148)
3-n-butylphthalideDrp-1/ROSIL-1βOGD(32)
Ketogenic DietDrp-1/ROS TXNIP/ATP NLRP3/IL-1β/Caspase-1MCAO/R OGD/R(147)
Panax ginseng and Angelica sinensisDrp-1 IL-1β/NLRP3/Caspase-1MCAO/R OGD/R(191)
FK866Mfn2/Drp1NLRP3/IL-1βCA/CPR(162)
isofluranemPTP/ROSIL-1βMCAO/OGD(192)
EzetimibeNrf2/TXNIP NLRP3/IL-1β/Caspase-1MCAO(171)
melatoninmtDNAIL-1βMCAO(193)
Necrostatin-1RIPK NLRP3/ASC/Caspase-1/IL-1βMCAO(140,141)
β-caryophylleneRIPKIL-1βOGD/R MCAO/R(194)
Isosteviol Sodium Ca2+/mtROSIL-1βhypoxia(195)
Taxifolin Ca2+IL-1βOGD(196)
Oxysophocarpine Ca2+IL-1βOGD/R(197)

[i] Focal cerebral ischemia was simulated through the MCAO in vivo and OGD/R in vitro. In MCAO and OGD/R models, drugs targeting specific targets of mitochondria and NLRP3 inflammasome for the treatment of ischemic stroke are listed. NLRP3, NLR family pyrin domain containing 3; ROS, reactive oxygen species; TXNIP, thioredoxin interacting protein; ASC, apoptosis-associated speck-like protein containing a CARD; MCAO, middle cerebral artery occlusion; OGD/R, oxygen-glucose deprivation/reoxygenation; tMCAO, transient MCAO; mtROS mitochondrial ROS; Drp1, dynamin-related protein 1; Mfn, mitofusin; RIPK, receptor-interacting protein kinase 1.

MCC950

As a sulfonylurea-containing compound, MCC950 was first identified as an IL-1β inhibitor (198). Then, MCC950 was also used as a specific inhibitor of NLRP3 inflammasome (199,200). The OGD/R-induced BV-2 cells and MCAO rats showed high expression of Drp1 and mitochondrial fission, as well as NLRP3 inflammasome activation, which were abolished by MCC950 treatments (191). Oxidative stress, mainly caused by mitochondria, was reported to be a crucial mechanism for brain damage following ischemic stroke. And NLRP3 inflammasome perpetuates oxidative stress. MCC950 application inhibits the effect of NLRP3 on brain oxidative stress in the animal model of transient global cerebral ischemia (201). In addition, MCC950 administration attenuated brain edema, reduced NADPH oxidase and infarct area and improved the nervous system in a MCAO rat model with reperfusion induced by hyperglycemia (202). Moreover, glibenclamide is a potent NLRP3 inflammasome inhibitor (203) that belongs to a class of medications known as sulfonylureas. Its neuroprotective role is due to its effect in reducing inflammatory response and endothelial cell death (204).

Idebenone

Idebenone was originally used as a drug to treat dementia. It was regarded as a potent antioxidant (205) which is used as a protective agent for mitochondria (206). Upon OGD/R, mtDNA and mtROS were released, resulting in accumulation of oxidized mtDNA in the cytoplasm which binds to and activates NLRP3 to initiate inflammation. Furthermore, idebenone treatment inhibited this process. NLRP3 was activated in microglia of ischemia-reperfusion rats in vivo. Inhibition of NLRP3 was observed in idebenone treatment which attenuated neurological deficit and reduced infarct volume (33).

Diazoxide

Diazoxide, an activator of mitochondrial K-ATP (207), prevents cytochrome c release and stabilizes mitochondrial function (208). Furthermore, diazoxide protects neurons during ischemia-reperfusion injury (209). Thus, diazoxide plays a crucial role in stabilizing mitochondrial function and in protecting neuronal survival. Indeed, mitochondrial dysfunction plays a pivotal role in the initiation and activation of the NLRP3 inflammasome (210). Diazoxide prohibits NLRP3 inflammasome activation to prevent inflammation (211). In addition, diazoxide improves mitochondrial dysfunction following ORG/R in primary microglia BV2 cell by preventing mitochondrial depolarization and the opening of MPTP to inhibit NLRP3 inflammasome activation (212). Therefore, maintaining mitochondrial stability and reducing NLRP3 inflammasome activation could both be potential targets of diazoxide to treat ischemic stroke.

Melatonin

Melatonin, an endogenous regulator, is a metabolite of tryptophan released from the pineal gland (213). It is involved in numerous physiological and pathophysiological processes including antioxidant (214), anti-apoptotic, and anti-inflammatory effects (215). Melatonin is of neuroprotective effect, which reduces infarct volume, lowers brain edema, and increases neurological scores. In addition, melatonin preserves mitochondrial membrane potential and mitochondrial complex I activity (216), and inhibits the opening of MPTPs and the abnormal release of cytochrome c (217), which is critical in reducing ischemia-reperfusion injury. Furthermore, melatonin is a relatively nontoxic molecule, which is safe to use in clinical trials. Previously, melatonin prevented IL-1β overexpression in the MCAO rat model (218). Additionally, Wang et al (219) showed that melatonin inhibited cell death, loss of mPTP, the release of mitochondrial factors, pro-IL-1β processing, and activation of caspase-1 induced by OGD. Furthermore, it decreased infarct size and improved neurological scores after MCAO in mice (219,220). Consequently, melatonin exerts neuroprotective and anti-inflammatory effects by modulating multiple targets in the NLRP3 inflammation (220).

Anthocyanins

Anthocyanins, which are effective flavonoid antioxidants (220) are natural plant pigments with a wide range of biological activities (221,222). Anthocyanins are flavylium-based multistates (223) and can reduce the damage to neurovascular unit in MCAO rats (224). Furthermore, anthocyanins inhibit NLRP3 expression in the brains of MCAO/R rats (225) and reduce cytosolic cytochrome c release to prevent apoptosis (226). Moreover, anthocyanins are neuroprotective in mouse model of pMCAO, decrease cerebral superoxide levels and inhibit AIF release from mitochondria (227). Cui et al (228) showed that purified anthocyanin extracts significantly reduce the expression of caspase-1 and NLRP3 and activate Nrf2 in the ischemia-reperfusion mice brain, thereby inhibiting inflammation and protecting the brain. Thus, anthocyanins may be a potential candidate for the prevention and treatment of stroke.

6. Clinical perspectives

At present, thrombolytic therapy is the only approved treatment for acute ischemic stroke (229), but only a minority of all patients with stroke are eligible for this treatment. At present, some drugs that target mitochondria and NLRP3 inflammasome are being tested in preclinical research. Idebenone, melatonin, minocycline and 3-n-butylphthalide were examined in clinical trials (Table II). It was demonstrated that no adverse events were reported as to the clinic usage of 3-n-butylphthalide (230). 3-n-butylphthalide showed favorable results and safety in the treatment of patients with moderate acute ischemic stroke (ClinicalTrials.gov Identifier: NCT02149875). Minocycline was reported to be safe and well tolerated with half-life of ~24 h. It may be an ideal drug to treat ischemic stroke when used together with tPA (ClinicalTrials.gov Identifier: NCT00630396) (231). In addition, the North Shore University Hospital is recruiting patients to study the effect of intra-arterial neuroprotective agents (minocycline) for recanalization of ischemic stroke (ClinicalTrials.gov Identifier: NCT05032781). Furthermore, a clinical trial is comparing the efficacy and safety of low-dose rivastigmine with ezetimibe and high-dose rivastigmine in the treatment of ischemic stroke (ClinicalTrials.gov Identifier: NCT03993236). In conclusion, drugs targeting mitochondria and NLRP3 inflammasome show promising therapeutic effects.

Table II

A summary of drugs in clinical trial data to treat ischemic stroke.

Table II

A summary of drugs in clinical trial data to treat ischemic stroke.

DrugTrial phaseTrial lengthPatients recruitedDosageMain resultsReference or trial identifier
IdebenonePhase 228 daysPatients with the A3243G mitochondrial DNA mutation and MELAS900 mg/day or 2,250 mg/dayHigh doses of idebenone can reduce venous and brain lactate concentrations and lighten fatigueNCT00887562
MelatoninPhase 414 daysAcute ischemic stroke14 mg/dailyNo results postedNCT01863277
MinocyclinePhase 1
Phase 2
3 monthsAcute ischemic strokeMinocycline was administered intravenously within 6 h of stroke symptom onset in preset dose tiers of 3, 4.5, 6, or 10 mg/kg daily over 72 hMinocycline may be an ideal agent to use with tissue plasminogen activatorNCT00630396
MinocyclinePhase 2
Phase 3
5 daysAcute ischemic strokeMinocycline will be administered either intravenously or orally once daily for 5 daysNo results postedNCT03320018
3-n-butylphthalidePhase 490 daysIschemic stroke25 mg butyphthalide and 0.9 mg/kg rtPA intravenously. Next day, butalbital 25 mg/day twice for 14 days, followed by butyphthalide capsules (0.2 g/day 3 times) orally for 90 daysNo results postedNCT03394950
3-n-butylphthalidePhase 1
Phase 2
10 daysAcute cerebral stroke Within 12 h for the first timeIntravenous infusion of 25 mg dl-3-n-butylphthalide twice daily for 10 daysThe 10-day treatment with NBP was found to be beneficial for the recovery of neurological and behavioral outcomes of patients with AISNCT02149875

[i] To clarify the current progress of drugs that may target mitochondria and NLRP3 inflammasome by describing the trial phase, trial length, dosage and final main effects of relevant drugs in clinical trials. NLRP3, NLR family pyrin domain containing 3.

7. Conclusion

Mitochondria are involved in various processes essential for cell survival, including energy production and physiological cell death mechanisms. Emerging knowledge about this organelle has shed light on its implication in inflammation. It is well accepted that post-ischemic neuroinflammation is one of the important mechanisms of ischemic brain injury. NLRP3 inflammasome has been found to play a key role in driving neuroinflammation in brain cells, such as cerebral microvascular endothelial cells, neurons and microglia during acute ischemic stroke. NLRP3 inflammasome was firstly activated in microglia and then expressed in microvascular endothelial cells and neurons of rat brains after ischemia-reperfusion injury. NLRP3 inflammasome can be activated by several factors, including the release of mitochondrial components, such as mtROS, cardiolipin, and mtDNA and mitochondrial related proteins, such as TXNIP and RIPK1 and some proteins that regulate the location of mitochondria. Through the present review, the close relationship between mitochondria and NLRP3 inflammasome and how mitochondrial damage contributes to ischemic damage by targeting neuroinflammation were discussed. Thus, maintaining mitochondrial homeostasis is important to ischemic stroke. Although the efficacy of tPA for acute ischemic stroke is well established, there are still serious side effects and limits. New therapeutic targets focusing on mitochondria, such as potential antioxidant or anti-inflammatory medicines, are promising therapeutic approaches in ischemic stroke. In addition, for ischemic stroke, some aforementioned drugs shall be administered in clinical trials currently recruiting patients, or are used in ongoing clinical trials or have been used in completed clinical trials. Thus, understanding the biology and regulation of inflammasome-mitochondria connections is required to treat ischemic stroke.

Availability of data and materials

Not applicable.

Authors' contributions

XLZ designed the review, prepared the tables and figures, and wrote the manuscript. WYZ were involved in the conception and design of the study. YZ and QY searched the literature. MZ, JLG and WLZ provided helpful comments. HHL were involved in the conception and design of the study and revised the manuscript. XJJ designed the study and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 82074211 and 82160828) and the Research and Innovation Project of Graduate (grant nos. 2020YJSS208, YJSKC-20201021 and ZXYCXLX201902).

Abbreviations:

ASC

apoptosis-associated speck-like protein containing a CARD

ARE

antioxidant response element

ANT1

adenine nucleotide translocase 1

DAMP

damage-associated molecular pattern

ER

endoplasmic reticulum

IL-1β

interleukin-1β

K-ATP

ATP-sensitive potassium channels

LRR

leucine rich repeat

mPTP

mitochondrial permeability transition pore

mtDNA

mitochondrial DNA

MCU

mitochondrial Ca2+ uniporter

MFN

mitofusin

MAM

mitochondria-associated ER membranes

mtROS

mitochondrial ROS

MRC

mitochondrial respiratory chain

NAD+

nicotinamide adenine dinucleotide

Nec-1

necrostatin-1

Nrf2

nuclear factor E2-related factor-2

OXPHOS

oxidative phosphorylation

OGD/R

oxygen-glucose deprivation/reoxygenation

PAMP

pathogen-associated molecular pattern

PTM

post-translational modification

RIPK1

receptor-interacting protein kinase 1

ROS

reactive oxygen species

SHP2

Src homology 2 domain-containing tyrosine phosphatase-2

TXNIP

thioredoxin interacting protein

TRX

thioredoxin

TRPM2

transient receptor potential melastain 2

tMCAO

transient middle cerebral artery occlusion

References

1 

Wang H, Wang Z, Wu Q, Yuan Y, Cao W and Zhang X: Regulatory T cells in ischemic stroke. CNS Neurosci Ther. 27:643–651. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Lu M, Guo J, Wu B, Zhou Y, Wu M, Farzaneh M and Khoshnam SE: Mesenchymal stem cell-mediated mitochondrial transfer: A therapeutic approach for ischemic stroke. Transl Stroke Res. 12:212–229. 2021. View Article : Google Scholar

3 

Feng L, Han CX, Cao SY, Zhang HM and Wu GY: Deficits in motor and cognitive functions in an adult mouse model of hypoxia-ischemia induced stroke. Sci Rep. 10:206462020. View Article : Google Scholar : PubMed/NCBI

4 

Barrington J, Lemarchand E and Allan SM: A brain in flame; do inflammasomes and pyroptosis influence stroke pathology? Brain Pathol. 27:205–212. 2017. View Article : Google Scholar

5 

Lambertsen KL, Finsen B and Clausen BH: Post-stroke inflammation-target or tool for therapy? Acta Neuropathol. 137:693–714. 2019. View Article : Google Scholar

6 

Andrabi SS, Parvez S and Tabassum H: Ischemic stroke and mitochondria: Mechanisms and targets. Protoplasma. 257:335–343. 2020. View Article : Google Scholar

7 

Li X, Huang Z, Liu S, Zeng X, Xie J, Liu C, Xiao H, Liu R, Li L and Zeng J: 3′-Daidzein sulfonate sodium provides neuroprotection by promoting the expression of the alpha7 nicotinic acetylcholine receptor and suppressing inflammatory responses in a rat model of focal cerebral ischemia. Am J Transl Res. 10:3455–3464. 2018.

8 

Mo Y, Sun YY and Liu KY: Autophagy and inflammation in ischemic stroke. Neural Regen Res. 15:1388–1396. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Jayaraj RL, Azimullah S, Beiram R, Jalal FY and Rosenberg GA: Neuroinflammation: Friend and foe for ischemic stroke. J Neuroinflammation. 16:1422019. View Article : Google Scholar : PubMed/NCBI

10 

Vats K, Sarmah D, Kaur H, Wanve M, Kalia K, Borah A, Dave KR, Yavagal DR and Bhattacharya P: Inflammasomes in stroke: A triggering role for acid-sensing ion channels. Ann N Y Acad Sci. 1431:14–24. 2018. View Article : Google Scholar

11 

Forn-Cuni G, Meijer AH and Varela M: Zebrafish in inflammasome research. Cells. 8:9012019. View Article : Google Scholar :

12 

Ma C, Liu S, Zhang S, Xu T, Yu X, Gao Y, Zhai C, Li C, Lei C, Fan S, et al: Evidence and perspective for the role of the NLRP3 inflammasome signaling pathway in ischemic stroke and its therapeutic potential (Review). Int J Mol Med. 42:2979–2990. 2018.

13 

Xu Q, Zhao B, Ye Y, Li Y, Zhang Y, Xiong X and Gu L: Relevant mediators involved in and therapies targeting the inflammatory response induced by activation of the NLRP3 inflammasome in ischemic stroke. J Neuroinflammation. 18:1232021. View Article : Google Scholar : PubMed/NCBI

14 

Qian Y, Lyu Y, Jiang M, Tang B, Nie T and Lu S: Human urinary kallidinogenase or edaravone combined with butylphthalide in the treatment of acute ischemic stroke. Brain Behav. 9:e014382019. View Article : Google Scholar : PubMed/NCBI

15 

Poh L, Kang SW, Baik SH, Ng GYQ, She DT, Balaganapathy P, Dheen ST, Magnus T, Gelderblom M, Sobey CG, et al: Evidence that NLRC4 inflammasome mediates apoptotic and pyroptotic microglial death following ischemic stroke. Brain Behav Immun. 75:34–47. 2019. View Article : Google Scholar

16 

Cao Y, Zhang H, Lu X, Wang J, Zhang X, Sun S, Bao Z, Tian W, Ning S, Wang L and Cui L: Overexpression of MicroRNA-9a-5p Ameliorates NLRP1 inflammasome-mediated ischemic injury in rats following ischemic stroke. Neuroscience. 444:106–117. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Xu SY, Bian HJ, Shu S, Xia SN, Gu Y, Zhang MJ, Xu Y and Cao X: AIM2 deletion enhances blood-brain barrier integrity in experimental ischemic stroke. CNS Neurosci Ther. 27:1224–1237. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Yu JW and Lee MS: Mitochondria and the NLRP3 inflammasome: Physiological and pathological relevance. Arch Pharm Res. 39:1503–1518. 2016. View Article : Google Scholar : PubMed/NCBI

19 

He Y, Hara H and Nunez G: Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 41:1012–1021. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Wang YL, Wu HR, Zhang SS, Xiao HL, Yu J, Ma YY, Zhang YD and Liu Q: Catalpol ameliorates depressive-like behaviors in CUMS mice via oxidative stress-mediated NLRP3 inflammasome and neuroinflammation. Transl Psychiatry. 11:3532021. View Article : Google Scholar : PubMed/NCBI

21 

Elliott EI and Sutterwala FS: Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol Rev. 265:35–52. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Savyuk M, Krivonosov M, Mishchenko T, Gazaryan I, Ivanchenko M, Khristichenko A, Poloznikov A, Hushpulian D, Nikulin S, Tonevitsky E, et al: Neuroprotective Effect of HIF prolyl hydroxylase inhibition in an in vitro hypoxia model. Antioxidants (Basel). 9:6622020. View Article : Google Scholar

23 

Huber W, Zanner R, Schneider G, Schmid R and Lahmer T: Assessment of regional perfusion and organ function: Less and non-invasive techniques. Front Med (Lausanne). 6:502019. View Article : Google Scholar

24 

Wang W, Zhao F, Ma X, Perry G and Zhu X: Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: Recent advances. Mol Neurodegener. 15:302020. View Article : Google Scholar : PubMed/NCBI

25 

Li W, Kui L, Demetrios T, Gong X and Tang M: A Glimmer of hope: Maintain mitochondrial homeostasis to mitigate Alzheimer's disease. Aging Dis. 11:1260–1275. 2020. View Article : Google Scholar :

26 

Ham PR and Raju R: Mitochondrial function in hypoxic ischemic injury and influence of aging. Prog Neurobiol. 157:92–116. 2017. View Article : Google Scholar

27 

Liu Y, Lin J, Wu X, Guo X, Sun H, Yu B, Shen J, Bai J, Chen Z, Yang H, et al: Aspirin-mediated attenuation of intervertebral disc degeneration by ameliorating reactive oxygen species in vivo and in vitro. Oxid Med Cell Longev. 2019:71898542019. View Article : Google Scholar

28 

Anzell AR, Maizy R, Przyklenk K and Sanderson TH: Mitochondrial quality control and disease: Insights into ischemia-reperfusion injury. Mol Neurobiol. 55:2547–2564. 2018. View Article : Google Scholar

29 

Babenko VA, Silachev DN, Popkov VA, Zorova LD, Pevzner IB, Plotnikov EY, Sukhikh GT and Zorov DB: Miro1 enhances mitochondria transfer from multipotent mesenchymal stem cells (MMSC) to neural cells and improves the efficacy of cell recovery. Molecules. 23:6872018. View Article : Google Scholar :

30 

Mondal NK, Behera J, Kelly KE, George AK, Tyagi PK and Tyagi N: Tetrahydrocurcumin epigenetically mitigates mitochondrial dysfunction in brain vasculature during ischemic stroke. Neurochem Int. 122:120–138. 2019. View Article : Google Scholar :

31 

Andrabi SS, Ali M, Tabassum H, Parveen S and Parvez S: Pramipexole prevents ischemic cell death via mitochondrial pathways in ischemic stroke. Dis Model Mech. 12:dmm0338602019. View Article : Google Scholar :

32 

Chen N, Zhou Z, Li J, Li B, Feng J, He D, Luo Y, Zheng X, Luo J and Zhang J: 3-n-butylphthalide exerts neuroprotective effects by enhancing anti-oxidation and attenuating mitochondrial dysfunction in an in vitro model of ischemic stroke. Drug Des Devel Ther. 12:4261–4271. 2018. View Article : Google Scholar :

33 

Peng J, Wang H, Gong Z, Li X, He L, Shen Q, Pan J and Peng Y: Idebenone attenuates cerebral inflammatory injury in ischemia and reperfusion via dampening NLRP3 inflammasome activity. Mol Immunol. 123:74–87. 2020. View Article : Google Scholar

34 

Luan Y, Yang D, Zhang Z, Bie X, Zhao H, Wang Y, Liu Y, Yang S, Zhou B, Xu Y, et al: Association study between genetic variation in whole mitochondrial genome and ischemic stroke. J Mol Neurosci. 71:2152–2162. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Sarmah D, Datta A, Raut S, Sarkar A, Shah B, Bohra M, Singh U, Jagtap P, Baidya F, Kalia K, et al: The role of inflammasomes in atherosclerosis and stroke pathogenesis. Curr Pharm Des. 26:4234–4245. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Martynov MY and Gusev EI: Current knowledge on the neuroprotective and neuroregenerative properties of citicoline in acute ischemic stroke. J Exp Pharmacol. 7:17–28. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Bissen D, Foss F and Acker-Palmer A: AMPA receptors and their minions: Auxiliary proteins in AMPA receptor trafficking. Cell Mol Life Sci. 76:2133–2169. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Chen Y, Qin C, Huang J, Tang X, Liu C, Huang K, Xu J, Guo G, Tong A and Zhou L: The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif. 53:e127812020. View Article : Google Scholar :

39 

Wen B, Xu K, Huang R, Jiang T, Wang J, Chen J, Chen J and He B: Preserving mitochondrial function by inhibiting GRP75 ameliorates neuron injury under ischemic stroke. Mol Med Rep. 25:1652022. View Article : Google Scholar : PubMed/NCBI

40 

Liu D, Gharavi R, Pitta M, Gleichmann M and Mattson MP: Nicotinamide prevents NAD+ depletion and protects neurons against excitotoxicity and cerebral ischemia: NAD+ consumption by SIRT1 may endanger energetically compromised neurons. Neuromolecular Med. 11:28–42. 2009. View Article : Google Scholar :

41 

Misawa T, Takahama M, Kozaki T, Lee H, Zou J, Saitoh T and Akira S: Microtubule-driven spatial arrangement of mitochondria promotes activation of the NLRP3 inflammasome. Nat Immunol. 14:454–460. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Li S, Wang T, Zhai L, Ge K, Zhao J, Cong W and Guo Y: Picroside II exerts a neuroprotective effect by inhibiting mPTP permeability and EndoG release after cerebral ischemia/reperfusion injury in rats. J Mol Neurosci. 64:144–155. 2018. View Article : Google Scholar

43 

Zheng W, Talley WL, Holstein DM, Wewer J and Lechleiter JD: P2Y1R-initiated, IP3R-dependent stimulation of astrocyte mitochondrial metabolism reduces and partially reverses ischemic neuronal damage in mouse. J Cereb Blood Flow Metab. 33:600–711. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Bonora M, Bononi A, De Marchi E, Giorgi C, Lebiedzinska M, Marchi S, Patergnani S, Rimessi A, Suski JM, Wojtala A, et al: Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle. 12:674–683. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, Ma Q, Tian F and Chen Y: Mff-Dependent mitochondrial fission contributes to the pathogenesis of cardiac microvasculature ischemia/reperfusion injury via induction of mROS-mediated cardiolipin oxidation and HK2/VDAC1 disassociation-involved mPTP opening. J Am Heart Assoc. 6:e0053282017. View Article : Google Scholar : PubMed/NCBI

46 

Jin X, Zhang J, An T, Zhao H, Fu W, Li D, Liu S, Cao X and Liu B: A Genome-wide screen in saccharomyces cerevisiae reveals a critical role for oxidative phosphorylation in cellular tolerance to lithium hexafluorophosphate. Cells. 10:8882021. View Article : Google Scholar :

47 

He J, Liu J, Huang Y, Zhuo Y, Chen W, Duan D, Tang X, Lu M and Hu Z: Olfactory mucosa mesenchymal stem cells alleviate cerebral ischemia/reperfusion injury via Golgi apparatus secretory pathway Ca2+-ATPase isoform1. Front Cell Dev Biol. 8:5865412020. View Article : Google Scholar

48 

Chen M, Wang M, Yang Q, Wang M, Wang Z, Zhu Y, Zhang Y, Wang C, Jia Y, Li Y and Wen A: Antioxidant effects of hydroxysafflor yellow A and acetyl-11-keto-β-boswellic acid in combination on isoproterenol-induced myocardial injury in rats. Int J Mol Med. 37:1501–1510. 2016. View Article : Google Scholar :

49 

Wang C, Hao J, Liu X, Li C, Yuan X, Lee RJ, Bai T and Wang D: Isoforsythiaside attenuates Alzheimer's disease via regulating mitochondrial function through the PI3K/AKT pathway. Int J Mol Sci. 21:56872020. View Article : Google Scholar

50 

Wang T, Wang F, Yu L and Li Z: Nobiletin alleviates cerebral ischemic-reperfusion injury via MAPK signaling pathway. Am J Transl Res. 11:5967–5977. 2019.PubMed/NCBI

51 

Zhao Q, Zhang C, Wang X, Chen L, Ji H and Zhang Y: (S)-ZJM-289, a nitric oxide-releasing derivative of 3-n-butylphthalide, protects against ischemic neuronal injury by attenuating mitochondrial dysfunction and associated cell death. Neurochem Int. 60:134–144. 2012. View Article : Google Scholar

52 

Tan YQ, Zhang X, Zhang S, Zhu T, Garg M, Lobie PE and Pandey V: Mitochondria: The metabolic switch of cellular oncogenic transformation. Biochim Biophys Acta Rev Cancer. 1876:1885342021. View Article : Google Scholar : PubMed/NCBI

53 

Jahani-Asl A, Cheung EC, Neuspiel M, MacLaurin JG, Fortin A, Park DS, McBride HM and Slack RS: Mitofusin 2 protects cerebellar granule neurons against injury-induced cell death. J Biol Chem. 282:23788–23798. 2007. View Article : Google Scholar : PubMed/NCBI

54 

McGahan L, Hakim AM and Robertson GS: Hippocampal Myc and p53 expression following transient global ischemia. Brain Res Mol Brain Res. 56:133–145. 1998. View Article : Google Scholar : PubMed/NCBI

55 

Li Y and Liu X: Novel insights into the role of mitochondrial fusion and fission in cardiomyocyte apoptosis induced by ischemia/reperfusion. J Cell Physiol. 233:5589–5597. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Grohm J, Kim SW, Mamrak U, Tobaben S, Cassidy-Stone A, Nunnari J, Plesnila N and Culmsee C: Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ. 19:1446–1458. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Zhao YX, Cui M, Chen SF, Dong Q and Liu XY: Amelioration of ischemic mitochondrial injury and Bax-dependent outer membrane permeabilization by Mdivi-1. CNS Neurosci Ther. 20:528–538. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Wang J, Xiong S, Xie C, Markesbery WR and Lovell MA: Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer's disease. J Neurochem. 93:953–962. 2005. View Article : Google Scholar : PubMed/NCBI

59 

West AP and Shadel GS: Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol. 17:363–375. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Guo S, Geng X, Lee H and Ding Y: Phenothiazine inhibits neuroinflammation and inflammasome activation independent of hypothermia after ischemic stroke. Mol Neurobiol. 58:6136–6152. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Lian L, Zhang Y, Liu L, Yang L, Cai Y, Zhang J and Xu S: Neuroinflammation in ischemic stroke: Focus on MicroRNA-mediated polarization of microglia. Front Mol Neurosci. 13:6124392020. View Article : Google Scholar

62 

Shaheryar ZA, Khan MA, Adnan CS, Zaidi AA, Hanggi D and Muhammad S: Neuroinflammatory triangle presenting novel pharmacological targets for ischemic brain injury. Front Immunol. 12:7486632021. View Article : Google Scholar : PubMed/NCBI

63 

Xue Y, Nie D, Wang LJ, Qiu HC, Ma L, Dong MX, Tu WJ and Zhao J: Microglial polarization: Novel therapeutic strategy against ischemic stroke. Aging Dis. 12:466–479. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Wang L, Yu CC, Liu XY, Deng XN, Tian Q and Du YJ: Epigenetic modulation of microglia function and phenotypes in neurodegenerative diseases. Neural Plast. 2021:99126862021. View Article : Google Scholar : PubMed/NCBI

65 

Ponsaerts L, Alders L, Schepers M, de Oliveira RMW, Prickaerts J, Vanmierlo T and Bronckaers A: Neuroinflammation in ischemic stroke: Inhibition of cAMP-Specific phosphodiesterases (PDEs) to the rescue. Biomedicines. 9:7032021. View Article : Google Scholar :

66 

Guan X, Zhang Y, Gareev I, Beylerli O, Li X, Lu X, Lv L and Hai X: MiR-499a prevents astrocytes mediated inflammation in ischemic stroke by targeting PTEN. Noncoding RNA Res. 6:146–152. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Franke M, Bieber M, Kraft P, Weber A, Stoll G and Schuhmann MK: The NLRP3 inflammasome drives inflammation in ischemia/reperfusion injury after transient middle cerebral artery occlusion in mice. Brain Behav Immun. 92:223–233. 2021. View Article : Google Scholar

68 

Gritsenko A, Green JP, Brough D and Lopez-Castejon G: Mechanisms of NLRP3 priming in inflammaging and age related diseases. Cytokine Growth Factor Rev. 55:15–25. 2020. View Article : Google Scholar : PubMed/NCBI

69 

Xu S, Li X, Liu Y, Xia Y, Chang R and Zhang C: Inflammasome inhibitors: Promising therapeutic approaches against cancer. J Hematol Oncol. 12:642019. View Article : Google Scholar : PubMed/NCBI

70 

Shim DW and Lee KH: Posttranslational regulation of the NLR family pyrin domain-containing 3 inflammasome. Front Immunol. 9:10542018. View Article : Google Scholar :

71 

Han S, Lear TB, Jerome JA, Rajbhandari S, Snavely CA, Gulick DL, Gibson KF, Zou C, Chen BB and Mallampalli RK: Lipopolysaccharide primes the NALP3 inflammasome by inhibiting its ubiquitination and degradation mediated by the SCFFBXL2 E3 ligase. J Biol Chem. 290:18124–18133. 2015. View Article : Google Scholar :

72 

Swanson KV, Deng M and Ting JP: The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat Rev Immunol. 19:477–489. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Susjan P, Roskar S and Hafner-Bratkovic I: The mechanism of NLRP3 inflammasome initiation: Trimerization but not dimerization of the NLRP3 pyrin domain induces robust activation of IL-1beta. Biochem Biophys Res Commun. 483:823–828. 2017. View Article : Google Scholar

74 

Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, Schröder GF, Fitzgerald KA, Wu H and Egelman EH: Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 156:1193–1206. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Boucher D, Monteleone M, Coll RC, Chen KW, Ross CM, Teo JL, Gomez GA, Holley CL, Bierschenk D, Stacey KJ, et al: Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity. J Exp Med. 215:827–840. 2018. View Article : Google Scholar :

76 

Dick MS, Sborgi L, Ruhl S, Hiller S and Broz P: ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun. 7:119292016. View Article : Google Scholar : PubMed/NCBI

77 

Lu L, Lu Q, Chen W, Li J, Li C and Zheng Z: Vitamin D3 protects against diabetic retinopathy by inhibiting high-glucose-induced activation of the ROS/TXNIP/NLRP3 inflammasome pathway. J Diabetes Res. 2018:81935232018. View Article : Google Scholar :

78 

Ratajczak MZ, Bujko K, Cymer M, Thapa A, Adamiak M, Ratajczak J, Abdel-Latif AK and Kucia M: The Nlrp3 inflammasome as a 'rising star' in studies of normal and malignant hematopoiesis. Leukemia. 34:1512–1523. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Gao L, Dong Q, Song Z, Shen F, Shi J and Li Y: NLRP3 inflammasome: A promising target in ischemic stroke. Inflamm Res. 66:17–24. 2017. View Article : Google Scholar

80 

Liu H, Wu X, Luo J, Zhao L, Li X, Guo H, Bai H, Cui W, Guo W, Feng D and Qu Y: Adiponectin peptide alleviates oxidative stress and NLRP3 inflammasome activation after cerebral ischemia-reperfusion injury by regulating AMPK/GSK-3beta. Exp Neurol. 329:1133022020. View Article : Google Scholar

81 

Zhao J, Piao X, Wu Y, Liang S, Han F, Liang Q, Shao S and Zhao D: Cepharanthine attenuates cerebral ischemia/reperfusion injury by reducing NLRP3 inflammasome-induced inflammation and oxidative stress via inhibiting 12/15-LOX signaling. Biomed Pharmacother. 127:1101512020. View Article : Google Scholar : PubMed/NCBI

82 

Yang F, Wang Z, Wei X, Han H, Meng X, Zhang Y, Shi W, Li F, Xin T, Pang Q and Yi F: NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab. 34:660–667. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Feng YS, Tan ZX, Wang MM, Xing Y, Dong F and Zhang F: Inhibition of NLRP3 inflammasome: A prospective target for the treatment of ischemic stroke. Front Cell Neurosci. 14:1552020. View Article : Google Scholar : PubMed/NCBI

84 

Shi M, Chen J, Liu T, Dai W, Zhou Z, Chen L and Xie Y: Protective effects of remimazolam on cerebral ischemia/reperfusion injury in rats by inhibiting of NLRP3 inflammasome-dependent pyroptosis. Drug Des Devel Ther. 16:413–423. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Ye Y, Jin T, Zhang X, Zeng Z, Ye B, Wang J, Zhong Y, Xiong X and Gu L: Meisoindigo protects against focal cerebral ischemia-reperfusion injury by inhibiting NLRP3 inflammasome activation and regulating microglia/macrophage polarization via TLR4/NF-κB signaling pathway. Front Cell Neurosci. 13:5532019. View Article : Google Scholar

86 

He Z, Ning N, Zhou Q, Khoshnam SE and Farzaneh M: Mitochondria as a therapeutic target for ischemic stroke. Free Radic Biol Med. 146:45–58. 2020. View Article : Google Scholar

87 

Chen Y, Zhou Z and Min W: Mitochondria, oxidative stress and innate immunity. Front Physiol. 9:14872018. View Article : Google Scholar : PubMed/NCBI

88 

Meyers AK and Zhu X: The NLRP3 inflammasome: Metabolic regulation and contribution to inflammaging. Cells. 9:18082020. View Article : Google Scholar :

89 

Bauernfeind F, Bartok E, Rieger A, Franchi L, Nunez G and Hornung V: Cutting edge: Reactive oxygen species inhibitors block priming, but not activation, of the NLRP3 inflammasome. J Immunol. 187:613–617. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Paik S, Kim JK, Silwal P, Sasakawa C and Jo EK: An update on the regulatory mechanisms of NLRP3 inflammasome activation. Cell Mol Immunol. 18:1141–1160. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Ren GM, Li J, Zhang XC, Wang Y, Xiao Y, Zhang XY, Liu X, Zhang W, Ma WB, Zhang J, et al: Pharmacological targeting of NLRP3 deubiquitination for treatment of NLRP3-associated inflammatory diseases. Sci Immunol. 6:eabe29332021. View Article : Google Scholar

92 

Ren JD, Wu XB, Jiang R, Hao DP and Liu Y: Molecular hydrogen inhibits lipopolysaccharide-triggered NLRP3 inflammasome activation in macrophages by targeting the mitochondrial reactive oxygen species. Biochim Biophys Acta. 1863:50–55. 2016. View Article : Google Scholar

93 

Wang Y, Shi P, Chen Q, Huang Z, Zou D, Zhang J, Gao X and Lin Z: Mitochondrial ROS promote macrophage pyroptosis by inducing GSDMD oxidation. J Mol Cell Biol. 11:1069–1082. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Heid ME, Keyel PA, Kamga C, Shiva S, Watkins SC and Salter RD: Mitochondrial reactive oxygen species induces NLRP3-dependent lysosomal damage and inflammasome activation. J Immunol. 191:5230–5238. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Qiu Z, He Y, Ming H, Lei S, Leng Y and Xia ZY: Lipopolysaccharide (LPS) aggravates high glucose- and hypoxia/reoxygenation-induced injury through activating ROS-Dependent NLRP3 inflammasome-mediated pyroptosis in H9C2 cardiomyocytes. J Diabetes Res. 2019:81518362019. View Article : Google Scholar : PubMed/NCBI

96 

Liu X, Zhang X, Ding Y, Zhou W, Tao L, Lu P, Wang Y and Hu R: Nuclear factor E2-Related Factor-2 negatively regulates NLRP3 inflammasome activity by inhibiting reactive oxygen species-induced NLRP3 priming. Antioxid Redox Signal. 26:28–43. 2017. View Article : Google Scholar :

97 

Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F and Alnemri ES: Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation. J Biol Chem. 287:36617–36622. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C, Lambrecht BN and Vandenabeele P: Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol. 32:157–164. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Arias-Cartin R, Grimaldi S, Arnoux P, Guigliarelli B and Magalon A: Cardiolipin binding in bacterial respiratory complexes: Structural and functional implications. Biochim Biophys Acta. 1817:1937–1949. 2012. View Article : Google Scholar : PubMed/NCBI

100 

Ji J, Baart S, Vikulina AS, Clark RS, Anthonymuthu TS, Tyurin VA, Du L, St Croix CM, Tyurina YY, Lewis J, et al: Deciphering of mitochondrial cardiolipin oxidative signaling in cerebral ischemia-reperfusion. J Cereb Blood Flow Metab. 35:319–328. 2015. View Article : Google Scholar :

101 

Liu J, Wang T, He K, Xu M and Gong JP: Cardiolipin inhibitor ameliorates the non-alcoholic steatohepatitis through suppressing NLRP3 inflammasome activation. Eur Rev Med Pharmacol Sci. 23:8158–8167. 2019.PubMed/NCBI

102 

Szeto HH, Liu S, Soong Y, Seshan SV, Cohen-Gould L, Manichev V, Feldman LC and Gustafsson T: Mitochondria protection after acute ischemia prevents prolonged upregulation of IL-1β and IL-18 and arrests CKD. J Am Soc Nephrol. 28:1437–1449. 2017. View Article : Google Scholar

103 

Carinci M, Vezzani B, Patergnani S, Ludewig P, Lessmann K, Magnus T, Casetta I, Pugliatti M, Pinton P and Giorgi C: Different roles of mitochondria in cell death and inflammation: Focusing on mitochondrial quality control in ischemic stroke and reperfusion. Biomedicines. 9:1692021. View Article : Google Scholar : PubMed/NCBI

104 

Yabal M, Calleja DJ, Simpson DS and Lawlor KE: Stressing out the mitochondria: Mechanistic insights into NLRP3 inflammasome activation. J Leukoc Biol. 105:377–399. 2019. View Article : Google Scholar

105 

Englander EW, Greeley GJ, Wang G, Perez-Polo JR and Lee HM: Hypoxia-induced mitochondrial and nuclear DNA damage in the rat brain. J Neurosci Res. 58:262–269. 1999. View Article : Google Scholar : PubMed/NCBI

106 

Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM, et al: Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 36:401–414. 2012. View Article : Google Scholar : PubMed/NCBI

107 

Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, et al: Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 12:222–2230. 2011. View Article : Google Scholar

108 

Lara PC, Macias-Verde D and Burgos-Burgos J: Age-induced NLRP3 inflammasome over-activation increases lethality of SARS-CoV-2 pneumonia in elderly patients. Aging Dis. 11:756–762. 2020. View Article : Google Scholar :

109 

Fu L, Zhang DX, Zhang LM, Song YC, Liu FH, Li Y, Wang XP, Zheng WC, Wang XD, Gui CX, et al: Exogenous carbon monoxide protects against mitochondrial DNAinduced hippocampal pyroptosis in a model of hemorrhagic shock and resuscitation. Int J Mol Med. 45:1176–1186. 2020.

110 

Simon R, Meller R, Yang T, Pearson A and Wilson G: Enhancing base excision repair of mitochondrial DNA to reduce ischemic injury following reperfusion. Transl Stroke Res. 10:664–671. 2019. View Article : Google Scholar :

111 

Gomez-Suaga P, Bravo-San PJ, Gonzalez-Polo RA, Fuentes JM and Niso-Santano M: ER-mitochondria signaling in Parkinson's disease. Cell Death Dis. 9:3372018. View Article : Google Scholar : PubMed/NCBI

112 

Tubbs E, Theurey P, Vial G, Bendridi N, Bravard A, Chauvin MA, Ji-Cao J, Zoulim F, Bartosch B, Ovize M, et al: Mitochondria-associated endoplasmic reticulum membrane (MAM) integrity is required for insulin signaling and is implicated in hepatic insulin resistance. Diabetes. 63:3279–3294. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Bravo R, Vicencio JM, Parra V, Troncoso R, Munoz JP, Bui M, Quiroga C, Rodriguez AE, Verdejo HE, Ferreira J, et al: Increased ER-mitochondrial coupling promotes mitochondrial respiration and bioenergetics during early phases of ER stress. J Cell Sci. 24:2143–2152. 2011. View Article : Google Scholar

114 

Elliott EI, Miller AN, Banoth B, Iyer SS, Stotland A, Weiss JP, Gottlieb RA, Sutterwala FS and Cassel SL: Cutting Edge: Mitochondrial assembly of the NLRP3 inflammasome complex is initiated at priming. J Immunol. 200:3047–3052. 2018. View Article : Google Scholar : PubMed/NCBI

115 

Hamilton C and Anand PK: Right place, right time: Localisation and assembly of the NLRP3 inflammasome. F1000Res. 8:F1000 Faculty Rev-676. 2019. View Article : Google Scholar : PubMed/NCBI

116 

Zhou R, Yazdi AS, Menu P and Tschopp J: A role for mitochondria in NLRP3 inflammasome activation. Nature. 469:221–225. 2011. View Article : Google Scholar

117 

Gu J, Zhang T, Guo J, Chen K, Li H and Wang J: PINK1 activation and translocation to mitochondria-associated membranes mediates mitophagy and protects against hepatic ischemia/reperfusion injury. Shock. 54:783–793. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Spescha RD, Klohs J, Semerano A, Giacalone G, Derungs RS, Reiner MF, Rodriguez Gutierrez D, Mendez-Carmona N, Glanzmann M, Savarese G, et al: Post-ischaemic silencing of p66Shc reduces ischaemia/reperfusion brain injury and its expression correlates to clinical outcome in stroke. Eur Heart J. 36:1590–1600. 2015. View Article : Google Scholar : PubMed/NCBI

119 

Thoudam T, Jeon JH, Ha CM and Lee IK: Role of Mitochondria-Associated endoplasmic reticulum membrane in inflammation-mediated metabolic diseases. Mediators Inflamm. 2016:18514202016. View Article : Google Scholar

120 

Fu MM and Holzbaur EL: Integrated regulation of motor-driven organelle transport by scaffolding proteins. Trends Cell Biol. 24:564–574. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Place DE and Kanneganti TD: Recent advances in inflammasome biology. Curr Opin Immunol. 50:32–38. 2018. View Article : Google Scholar :

122 

Harkcom WT, Ghosh AK, Sung MS, Matov A, Brown KD, Giannakakou P and Jaffrey SR: NAD+ and SIRT3 control microtubule dynamics and reduce susceptibility to antimicrotubule agents. Proc Natl Acad Sci USA. 111:E2443–E2452. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Nasoohi S, Ismael S and Ishrat T: Thioredoxin-Interacting Protein (TXNIP) in Cerebrovascular and Neurodegenerative Diseases: Regulation and Implication. Mol Neurobiol. 55:7900–7920. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Nagaraj K, Lapkina-Gendler L, Sarfstein R, Gurwitz D, Pasmanik-Chor M, Laron Z, Yakar S and Werner H: Identification of thioredoxin-interacting protein (TXNIP) as a downstream target for IGF1 action. Proc Natl Acad Sci USA. 115:1045–1050. 2018. View Article : Google Scholar : PubMed/NCBI

125 

Zhou R, Tardivel A, Thorens B, Choi I and Tschopp J: Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 11:136–140. 2010. View Article : Google Scholar

126 

Fann DY, Lee SY, Manzanero S, Chunduri P, Sobey CG and Arumugam TV: Pathogenesis of acute stroke and the role of inflammasomes. Ageing Res Rev. 12:941–966. 2013. View Article : Google Scholar : PubMed/NCBI

127 

Han Y, Xu X, Tang C, Gao P, Chen X, Xiong X, Yang M, Yang S, Zhu X, Yuan S, et al: Reactive oxygen species promote tubular injury in diabetic nephropathy: The role of the mitochondrial ros-txnip-nlrp3 biological axis. Redox Biol. 16:32–46. 2018. View Article : Google Scholar : PubMed/NCBI

128 

Wang BF and Yoshioka J: The Emerging role of thioredoxin-interacting protein in myocardial ischemia/reperfusion injury. J Cardiovasc Pharmacol Ther. 22:219–229. 2017. View Article : Google Scholar :

129 

Schafer MK, Pfeiffer A, Jaeckel M, Pouya A, Dolga AM and Methner A: Regulators of mitochondrial Ca(2+) homeostasis in cerebral ischemia. Cell Tissue Res. 357:395–405. 2014. View Article : Google Scholar

130 

Missiroli S, Patergnani S, Caroccia N, Pedriali G, Perrone M, Previati M, Wieckowski MR and Giorgi C: Mitochondria-associated membranes (MAMs) and inflammation. Cell Death Dis. 9:3292018. View Article : Google Scholar : PubMed/NCBI

131 

Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, Germain RN, Kastner DL and Chae JJ: The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature. 492:123–127. 2012. View Article : Google Scholar : PubMed/NCBI

132 

Wang C, Jia Q, Sun C and Jing C: Calcium sensing receptor contribute to early brain injury through the CaMKII/NLRP3 pathway after subarachnoid hemorrhage in mice. Biochem Biophys Res Commun. 530:651–657. 2020. View Article : Google Scholar : PubMed/NCBI

133 

Triantafilou K, Hughes TR, Triantafilou M and Morgan BP: The complement membrane attack complex triggers intracellular Ca2+ fluxes leading to NLRP3 inflammasome activation. J Cell Sci. 126:2903–2913. 2013.PubMed/NCBI

134 

Murakami T, Ockinger J, Yu J, Byles V, McColl A, Hofer AM and Horng T: Critical role for calcium mobilization in activation of the NLRP3 inflammasome. Proc Natl Acad Sci USA. 109:11282–11287. 2012. View Article : Google Scholar : PubMed/NCBI

135 

Pan T, Zhu QJ, Xu LX, Ding X, Li JQ, Sun B, Hua J and Feng X: Knocking down TRPM2 expression reduces cell injury and NLRP3 inflammasome activation in PC12 cells subjected to oxygen-glucose deprivation. Neural Regen Res. 15:2154–2161. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Chen AQ, Fang Z, Chen XL, Yang S, Zhou YF, Mao L, Xia YP, Jin HJ, Li YN, You MF, et al: Microglia-derived TNF-α mediates endothelial necroptosis aggravating blood brain-barrier disruption after ischemic stroke. Cell Death Dis. 10:4872019. View Article : Google Scholar

137 

Wang H, Sun L, Su L, Rizo J, Liu L, Wang LF, Wang FS and Wang X: Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol Cell. 54:133–146. 2014. View Article : Google Scholar : PubMed/NCBI

138 

He S and Wang X: RIP kinases as modulators of inflammation and immunity. Nat Immunol. 19:912–922. 2018. View Article : Google Scholar : PubMed/NCBI

139 

Zeng F, Chen X, Cui W, Wen W, Lu F, Sun X, Ma D, Yuan Y, Li Z, Hou N, et al: RIPK1 Binds MCU to mediate induction of mitochondrial Ca2+ uptake and promotes colorectal oncogenesis. Cancer Res. 78:2876–2885. 2018. View Article : Google Scholar : PubMed/NCBI

140 

Jiao Y, Wang J, Zhang H, Cao Y, Qu Y, Huang S, Kong X, Song C, Li J, Li Q, et al: Inhibition of microglial receptor-interacting protein kinase 1 ameliorates neuroinflammation following cerebral ischaemic stroke. J Cell Mol Med. 24:12585–12598. 2020. View Article : Google Scholar : PubMed/NCBI

141 

Deng XX, Li SS and Sun FY: Necrostatin-1 prevents necroptosis in brains after ischemic stroke via inhibition of RIPK1-Mediated RIPK3/MLKL signaling. Aging Dis. 10:807–817. 2019. View Article : Google Scholar : PubMed/NCBI

142 

Park S, Won JH, Hwang I, Hong S, Lee HK and Yu JW: Defective mitochondrial fission augments NLRP3 inflammasome activation. Sci Rep. 5:154892015. View Article : Google Scholar : PubMed/NCBI

143 

Xie JH, Li YY and Jin J: The essential functions of mitochondrial dynamics in immune cells. Cell Mol Immunol. 17:712–721. 2020. View Article : Google Scholar : PubMed/NCBI

144 

Ren L, Chen X, Chen X, Li J, Cheng B and Xia J: Mitochondrial dynamics: Fission and fusion in fate determination of mesenchymal stem cells. Front Cell Dev Biol. 8:5800702020. View Article : Google Scholar : PubMed/NCBI

145 

Szabadkai G, Simoni AM, Chami M, Wieckowski MR, Youle RJ and Rizzuto R: Drp-1-dependent division of the mitochondrial network blocks intraorganellar Ca2+ waves and protects against Ca2+-mediated apoptosis. Mol Cell. 16:59–68. 2004. View Article : Google Scholar : PubMed/NCBI

146 

Flippo KH, Gnanasekaran A, Perkins GA, Ajmal A, Merrill RA, Dickey AS, Taylor SS, McKnight GS, Chauhan AK, Usachev YM and Strack S: AKAP1 protects from cerebral ischemic stroke by inhibiting Drp1-dependent mitochondrial fission. J Neurosci. 38:8233–8242. 2018. View Article : Google Scholar : PubMed/NCBI

147 

Guo M, Wang X, Zhao Y, Yang Q, Ding H, Dong Q, Chen X and Cui M: Ketogenic diet improves brain ischemic tolerance and inhibits NLRP3 inflammasome activation by preventing Drp1-Mediated mitochondrial fission and endoplasmic reticulum stress. Front Mol Neurosci. 11:862018. View Article : Google Scholar :

148 

He J and Zhang X: miR-668 inhibitor attenuates mitochondrial membrane potential and protects against neuronal apoptosis in cerebral ischemic stroke. Folia Neuropathol. 58:22–29. 2020. View Article : Google Scholar

149 

Zhang X, Yan H, Yuan Y, Gao J, Shen Z, Cheng Y, Shen Y, Wang RR, Wang X, Hu WW, et al: Cerebral ischemia-reperfusion-induced autophagy protects against neuronal injury by mitochondrial clearance. Autophagy. 9:1321–1333. 2013. View Article : Google Scholar : PubMed/NCBI

150 

Wang J, Yu S, Li J, Li H, Jiang H, Xiao P, Pan Y, Zheng J, Yu L and Jiang J: Protective role of N-acetyl-l-tryptophan against hepatic ischemia-reperfusion injury via the RIP2/caspase-1/IL-1beta signaling pathway. Pharm Biol. 57:385–391. 2019. View Article : Google Scholar : PubMed/NCBI

151 

Wang Y, Tian J, Qiao X, Su X, Mi Y, Zhang R and Li R: Intermedin protects against renal ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress. BMC Nephrol. 16:1692015. View Article : Google Scholar : PubMed/NCBI

152 

He Q, Li Z, Meng C, Wu J, Zhao Y and Zhao J: Parkin-dependent mitophagy is required for the inhibition of ATF4 on NLRP3 inflammasome activation in cerebral ischemia-reperfusion injury in rats. Cells. 8:8972019. View Article : Google Scholar :

153 

Yang J, Chen Y and Pang Y: Occurrence of mitochondrial autophagy and nlrp3 inflammatory bodies in cerebral ischemia-reperfusion injury and its correlation with neuroinflammatory response. Acta Medica Mediterranea. 37:1033–1037. 2021.

154 

Su SH, Wu YF, Lin Q, Wang DP and Hai J: URB597 protects against NLRP3 inflammasome activation by inhibiting autophagy dysfunction in a rat model of chronic cerebral hypoperfusion. J Neuroinflammation. 16:2602019. View Article : Google Scholar : PubMed/NCBI

155 

Fan Y, Zhu S, Wang J, Zhao Y and Wang X: Propofol protects against oxygen/glucose deprivationinduced cell injury via gap junction inhibition in astrocytes. Mol Med Rep. 22:2896–2904. 2020.PubMed/NCBI

156 

Cai Y, Guo H, Fan Z, Zhang X, Wu D, Tang W, Gu T, Wang S, Yin A, Tao L, et al: Glycogenolysis is crucial for astrocytic glycogen accumulation and brain damage after reperfusion in ischemic stroke. iScience. 23:1011362020. View Article : Google Scholar : PubMed/NCBI

157 

Gao L, Liu F, Hou PP, Manaenko A, Xiao ZP, Wang F, Xu TL and Hu Q: Neurons release injured mitochondria as 'Help-Me' signaling after ischemic stroke. Front Aging Neurosci. 14:7857612022. View Article : Google Scholar

158 

Jiang D, Gao F, Zhang Y, Wong DS, Li Q, Tse HF, Xu G, Yu Z and Lian Q: Mitochondrial transfer of mesenchymal stem cells effectively protects corneal epithelial cells from mitochondrial damage. Cell Death Dis. 7:e24672016. View Article : Google Scholar : PubMed/NCBI

159 

Hasan-Olive MM, Enger R, Hansson HA, Nagelhus EA and Eide PK: Pathological mitochondria in neurons and perivascular astrocytic endfeet of idiopathic normal pressure hydrocephalus patients. Fluids Barriers CNS. 16:392019. View Article : Google Scholar :

160 

Guo W, Liu W, Chen Z, Gu Y, Peng S, Shen L, Shen Y, Wang X, Feng GS, Sun Y and Xu Q: Tyrosine phosphatase SHP2 negatively regulates NLRP3 inflammasome activation via ANT1-dependent mitochondrial homeostasis. Nat Commun. 8:21682017. View Article : Google Scholar :

161 

Aoki Y, Huang Z, Thomas SS, Bhide PG, Huang I, Moskowitz MA and Reeves SA: Increased susceptibility to ischemia-induced brain damage in transgenic mice overexpressing a dominant negative form of SHP2. FASEB J. 14:1965–1973. 2000. View Article : Google Scholar : PubMed/NCBI

162 

Zou X, Xie L, Wang W, Zhao G, Tian X and Chen M: FK866 alleviates cerebral pyroptosis and inflammation mediated by Drp1 in a rat cardiopulmonary resuscitation model. Int Immunopharmacol. 89:1070322020. View Article : Google Scholar : PubMed/NCBI

163 

Dong J, Bobe G, Guan Y, Li G, Zuo R, Shu X, Wang Y, Sun X, Chen X and Li X: Mitochondrial membrane protein mitofusin 2 as a potential therapeutic target for treating free fatty acid-induced hepatic inflammation in dairy cows during early lactation. J Dairy Sci. 103:5561–5574. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Peng C, Rao W, Zhang L, Wang K, Hui H, Wang L, Su N, Luo P, Hao YL, Tu Y, et al: Mitofusin 2 ameliorates hypoxia-induced apoptosis via mitochondrial function and signaling pathways. Int J Biochem Cell Biol. 69:29–40. 2015. View Article : Google Scholar : PubMed/NCBI

165 

Wang X, Chen T, Ma X, Huang W, Huang Q, Liu K and Liang H: Progress on nuclear factor-E2 related factor 2 transcription factors in sepsis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 30:810–814. 2018.In Chinese. PubMed/NCBI

166 

Zhao C, Gillette DD, Li X, Zhang Z and Wen H: Nuclear factor E2-related factor-2 (Nrf2) is required for NLRP3 and AIM2 inflammasome activation. J Biol Chem. 289:17020–17029. 2014. View Article : Google Scholar : PubMed/NCBI

167 

Anandhan A, Nguyen N, Syal A, Dreher LA, Dodson M, Zhang DD and Madhavan L: NRF2 loss accentuates Parkinsonian pathology and behavioral dysfunction in human α-synuclein overexpressing mice. Aging Dis. 12:964–982. 2021. View Article : Google Scholar : PubMed/NCBI

168 

Li W, Khor TO, Xu C, Shen G, Jeong WS, Yu S and Kong AN: Activation of Nrf2-antioxidant signaling attenuates NFkappaB-inflammatory response and elicits apoptosis. Biochem Pharmacol. 76:1485–1489. 2008. View Article : Google Scholar

169 

Xu X, Zhang L, Ye X, Hao Q, Zhang T, Cui G and Yu M: Nrf2/ARE pathway inhibits ROS-induced NLRP3 inflammasome activation in BV2 cells after cerebral ischemia reperfusion. Inflamm Res. 67:57–65. 2018. View Article : Google Scholar

170 

Xu B, Zhang J, Strom J, Lee S and Chen QM: Myocardial ischemic reperfusion induces de novo Nrf2 protein translation. Biochim Biophys Acta. 1842:1638–1647. 2014. View Article : Google Scholar : PubMed/NCBI

171 

Yu J, Wang WN, Matei N, Li X, Pang JW, Mo J, Chen SP, Tang JP, Yan M and Zhang JH: Ezetimibe attenuates oxidative stress and neuroinflammation via the AMPK/Nrf2/TXNIP Pathway after MCAO in Rats. Oxid Med Cell Longev. 2020:47172582020. View Article : Google Scholar : PubMed/NCBI

172 

Hou Y, Wang Y, He Q, Li L, Xie H, Zhao Y and Zhao J: Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury. Behav Brain Res. 336:32–39. 2018. View Article : Google Scholar

173 

Zhang C, He M, Ni L, He K, Su K, Deng Y, Li Y and Xia H: The role of arachidonic acid metabolism in myocardial ischemia-reperfusion injury. Cell Biochem Biophys. 78:255–265. 2020. View Article : Google Scholar : PubMed/NCBI

174 

Shi Y, Peng XH, Li X, Luo GP and Wu MF: Neuroprotective role of dexmedetomidine pretreatment in cerebral ischemia injury via ADRA2A-mediated phosphorylation of ERK1/2 in adult rats. Exp Ther Med. 16:5201–5209. 2018.PubMed/NCBI

175 

Liu F, Lu J, Manaenko A, Tang J and Hu Q: Mitochondria in Ischemic Stroke: New Insight and Implications. Aging Dis. 9:924–937. 2018. View Article : Google Scholar : PubMed/NCBI

176 

Wang X, Li R, Wang X, Fu Q and Ma S: Umbelliferone ameliorates cerebral ischemia-reperfusion injury via upregulating the PPAR gamma expression and suppressing TXNIP/NLRP3 inflammasome. Neurosci Lett. 600:182–187. 2015. View Article : Google Scholar : PubMed/NCBI

177 

Li Y, Li J, Li S, Li Y, Wang X, Liu B, Fu Q and Ma S: Curcumin attenuates glutamate neurotoxicity in the hippocampus by suppression of ER stress-associated TXNIP/NLRP3 inflammasome activation in a manner dependent on AMPK. Toxicol Appl Pharmacol. 286:53–63. 2015. View Article : Google Scholar : PubMed/NCBI

178 

Cao G, Jiang N, Hu Y, Zhang Y, Wang G, Yin M, Ma X, Zhou K, Qi J, Yu B and Kou J: Ruscogenin attenuates cerebral ischemia-induced blood-brain barrier dysfunction by suppressing TXNIP/NLRP3 inflammasome activation and the MAPK pathway. Int J Mol Sci. 17:14182016. View Article : Google Scholar :

179 

Ishrat T, Mohamed IN, Pillai B, Soliman S, Fouda AY, Ergul A, El-Remessy AB and Fagan SC: Thioredoxin-interacting protein: A novel target for neuroprotection in experimental thromboembolic stroke in mice. Mol Neurobiol. 51:766–778. 2015. View Article : Google Scholar

180 

Ismael S, Nasoohi S, Yoo A, Ahmed HA and Ishrat T: Tissue plasminogen activator promotes TXNIP-NLRP3 inflammasome activation after hyperglycemic stroke in mice. Mol Neurobiol. 57:2495–2508. 2020. View Article : Google Scholar : PubMed/NCBI

181 

Liu T, Wang W, Liu M, Ma Y, Mu F, Feng X, Zhang Y, Guo C, Ding Y and Wen A: Z-Guggulsterone alleviated oxidative stress and inflammation through inhibiting the TXNIP/NLRP3 axis in ischemic stroke. Int Immunopharmacol. 89:1070942020. View Article : Google Scholar : PubMed/NCBI

182 

Yang W, Chen X, Pan J, Ge H, Yin K, Wu Z, Li X, Sha D and Xu Y: Malibatol A protects against brain injury through reversing mitochondrial dysfunction in experimental stroke. Neurochem Int. 80:33–40. 2015. View Article : Google Scholar

183 

Gao XJ, Xie GN, Liu L, Fu ZJ, Zhang ZW and Teng LZ: Sesamol attenuates oxidative stress, apoptosis and inflammation in focal cerebral ischemia/reperfusion injury. Exp Ther Med. 14:841–847. 2017. View Article : Google Scholar : PubMed/NCBI

184 

Lu Y, Xiao G and Luo W: Minocycline suppresses NLRP3 inflammasome activation in experimental ischemic stroke. Neuroimmunomodulation. 23:230–238. 2016. View Article : Google Scholar : PubMed/NCBI

185 

Qiu J, Wang M, Zhang J, Cai Q, Lu D, Li Y, Dong Y, Zhao T and Chen H: The neuroprotection of Sinomenine against ischemic stroke in mice by suppressing NLRP3 inflammasome via AMPK signaling. Int Immunopharmacol. 40:492–500. 2016. View Article : Google Scholar : PubMed/NCBI

186 

Peng J, Deng X, Huang W, Yu JH, Wang JX, Wang JP, Yang SB, Liu X, Wang L, Zhang Y, et al: Irisin protects against neuronal injury induced by oxygen-glucose deprivation in part depends on the inhibition of ROS-NLRP3 inflammatory signaling pathway. Mol Immunol. 91:185–194. 2017. View Article : Google Scholar : PubMed/NCBI

187 

Qin YY, Li M, Feng X, Wang J, Cao L, Shen XK, Chen J, Sun M, Sheng R, Han F and Qin ZH: Combined NADPH and the NOX inhibitor apocynin provides greater anti-inflammatory and neuroprotective effects in a mouse model of stroke. Free Radic Biol Med. 104:333–345. 2017. View Article : Google Scholar : PubMed/NCBI

188 

Safakheil M and Safakheil H: The effect of exosomes derived from bone marrow stem cells in combination with rosuvastatin on functional recovery and neuroprotection in rats after ischemic stroke. J Mol Neurosci. 70:724–737. 2020. View Article : Google Scholar : PubMed/NCBI

189 

Barakat W, Fahmy A, Askar M and El-Kannishy S: Effectiveness of arginase inhibitors against experimentally induced stroke. Naunyn Schmiedebergs Arch Pharmacol. 391:603–612. 2018. View Article : Google Scholar : PubMed/NCBI

190 

Wang Y, Guan X, Gao CL, Ruan W, Zhao S, Kai G, Li F and Pang T: Medioresinol as a novel PGC-1α activator prevents pyroptosis of endothelial cells in ischemic stroke through PPARα-GOT1 axis. Pharmacol Res. 169:1056402021. View Article : Google Scholar

191 

Hu J, Zeng C, Wei J, Duan F, Liu S, Zhao Y and Tan H: The combination of Panax ginseng and Angelica sinensis alleviates ischemia brain injury by suppressing NLRP3 inflammasome activation and microglial pyroptosis. Phytomedicine. 76:1532512020. View Article : Google Scholar : PubMed/NCBI

192 

Yao Z, Liu N, Zhu X, Wang L, Zhao Y, Liu Q, Gao C and Li J: Subanesthetic isoflurane abates ROS-activated MAPK/NF-κB signaling to repress ischemia-induced microglia inflammation and brain injury. Aging (Albany NY). 12:26121–26139. 2020. View Article : Google Scholar

193 

Lin KC, Chen KH, Wallace CG, Chen YL, Ko SF, Lee MS and Yip HK: Combined therapy with hyperbaric oxygen and melatonin effectively reduce brain infarct volume and preserve neurological function after acute ischemic infarct in rat. J Neuropathol Exp Neurol. 78:949–960. 2019. View Article : Google Scholar : PubMed/NCBI

194 

Yang M, Lv Y, Tian X, Lou J, An R, Zhang Q, Li M, Xu L and Dong Z: Neuroprotective effect of β-caryophyllene on cerebral ischemia-reperfusion injury via regulation of necroptotic neuronal death and inflammation: In vivo and in vitro. Front Neurosci. 11:5832017. View Article : Google Scholar

195 

Zhong KL, Lu MY, Liu F, Mei Y, Zhang XJ, Zhang H, Zan J, Sun XO and Tan W: Isosteviol sodium protects neural cells against hypoxia-induced apoptosis through inhibiting MAPK and NF-κB pathways. J Stroke Cerebrovasc Dis. 28:175–184. 2019. View Article : Google Scholar

196 

Turovskaya MV, Gaidin SG, Mal'Tseva VN, Zinchenko VP and Turovsky EA: Taxifolin protects neurons against ischemic injury in vitro via the activation of antioxidant systems and signal transduction pathways of GABAergic neurons. Mol Cell Neurosci. 96:10–24. 2019. View Article : Google Scholar : PubMed/NCBI

197 

Zhao P, Chang RY, Liu N, Wang J, Zhou R, Qi X, Liu Y, Ma L, Niu Y, Sun T, et al: Neuroprotective effect of oxysophocarpine by modulation of MAPK pathway in rat hippocampal neurons subject to oxygen-glucose deprivation and reperfusion. Cell Mol Neurobiol. 38:529–540. 2018. View Article : Google Scholar

198 

Wu D, Chen Y, Sun Y, Gao Q, Li H, Yang Z, Wang Y, Jiang X and Yu B: Target of MCC950 in Inhibition of NLRP3 inflammasome activation: A literature review. Inflammation. 43:17–23. 2020. View Article : Google Scholar

199 

Coll RC, Hill JR, Day CJ, Zamoshnikova A, Boucher D, Massey NL, Chitty JL, Fraser JA, Jennings MP, Robertson AAB and Schroder K: MCC950 directly targets the NLRP3 ATP-hydrolysis motif for inflammasome inhibition. Nat Chem Biol. 15:556–559. 2019. View Article : Google Scholar : PubMed/NCBI

200 

Jiao J, Zhao G, Wang Y, Ren P and Wu M: MCC950, a selective inhibitor of NLRP3 inflammasome, reduces the inflammatory response and improves neurological outcomes in mice model of spinal cord injury. Front Mol Biosci. 7:372020. View Article : Google Scholar : PubMed/NCBI

201 

Joaquim LS, Danielski LG, Bonfante S, Biehl E, Mathias K, Denicol T, Bagio E, Lanzzarin EV, Machado RS, Bernades GC, et al: NLRP3 inflammasome activation increases brain oxidative stress after transient global cerebral ischemia in rats. Int J Neurosci. 1–14. 2021.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

202 

Chen H, Guan B, Chen S, Yang D and Shen J: Peroxynitrite activates NLRP3 inflammasome and contributes to hemorrhagic transformation and poor outcome in ischemic stroke with hyperglycemia. Free Radic Biol Med. 165:171–183. 2021. View Article : Google Scholar : PubMed/NCBI

203 

Dwivedi DK and Jena GB: NLRP3 inhibitor glibenclamide attenuates high-fat diet and streptozotocin-induced non-alcoholic fatty liver disease in rat: Studies on oxidative stress, inflammation, DNA damage and insulin signalling pathway. Naunyn Schmiedebergs Arch Pharmacol. 393:705–716. 2020. View Article : Google Scholar

204 

Zhu S, Gao X, Huang K, Gu Y, Hu Y, Wu Y, Ji Z, Wang Q and Pan S: Glibenclamide enhances the therapeutic benefits of early hypothermia after severe stroke in rats. Aging Dis. 9:685–695. 2018. View Article : Google Scholar : PubMed/NCBI

205 

Gueven N, Ravishankar P, Eri R and Rybalka E: Idebenone: When an antioxidant is not an antioxidant. Redox Biol. 38:1018122021. View Article : Google Scholar

206 

Jiang W, Geng H, Lv X, Ma J, Liu F, Lin P and Yan C: Idebenone Protects against atherosclerosis in apolipoprotein E-Deficient mice via activation of the SIRT3-SOD2-mtROS pathway. Cardiovasc Drugs Ther. 35:1129–1145. 2021. View Article : Google Scholar

207 

Akopova O, Kolchinskaya L, Nosar V, Mankovska I and Sagach V: Diazoxide affects mitochondrial bioenergetics by the opening of mKATP channel on submicromolar scale. BMC Mol Cell Biol. 21:312020. View Article : Google Scholar : PubMed/NCBI

208 

Liu D, Lu C, Wan R, Auyeung WW and Mattson MP: Activation of mitochondrial ATP-dependent potassium channels protects neurons against ischemia-induced death by a mechanism involving suppression of Bax translocation and cytochrome c release. J Cereb Blood Flow Metab. 22:431–443. 2002. View Article : Google Scholar : PubMed/NCBI

209 

Lei X, Lei L, Zhang Z and Cheng Y: Diazoxide inhibits of ER stressmediated apoptosis during oxygenglucose deprivation in vitro and cerebral ischemiareperfusion in vivo. Mol Med Rep. 17:8039–8046. 2018.PubMed/NCBI

210 

Mishra SR, Mahapatra KK, Behera BP, Patra S, Bhol CS, Panigrahi DP, Praharaj PP, Singh A, Patil S, Dhiman R and Bhutia SK: Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 136:1060132021. View Article : Google Scholar : PubMed/NCBI

211 

Nógrádi B, Nyúl-Tóth Á, Kozma M, Molnár K, Patai R, Siklós L, Wilhelm I and Krizbai IA: Upregulation of nucleotide-binding oligomerization Domain-, LRR- and pyrin domain-containing protein 3 in motoneurons following peripheral nerve injury in mice. Front Pharmacol. 11:5841842020. View Article : Google Scholar : PubMed/NCBI

212 

Gong Z, Pan J, Shen Q, Li M and Peng Y: Mitochondrial dysfunction induces NLRP3 inflammasome activation during cerebral ischemia/reperfusion injury. J Neuroinflammation. 15:2422018. View Article : Google Scholar : PubMed/NCBI

213 

Kondoh T, Uneyama H, Nishino H and Torii K: Melatonin reduces cerebral edema formation caused by transient forebrain ischemia in rats. Life Sci. 72:583–590. 2002. View Article : Google Scholar : PubMed/NCBI

214 

Kilic E, Caglayan B and Caglar BM: Physiological and pharmacological roles of melatonin in the pathophysiological components of cellular injury after ischemic stroke. Turk J Med Sci. 50:1655–1664. 2020. View Article : Google Scholar : PubMed/NCBI

215 

Fan W, He Y, Guan X, Gu W, Wu Z, Zhu X, Huang F and He H: Involvement of the nitric oxide in melatonin-mediated protection against injury. Life Sci. 200:142–147. 2018. View Article : Google Scholar : PubMed/NCBI

216 

Yang Y, Jiang S, Dong Y, Fan C, Zhao L, Yang X, Li J, Di S, Yue L, Liang G, et al: Melatonin prevents cell death and mitochondrial dysfunction via a SIRT1-dependent mechanism during ischemic-stroke in mice. J Pineal Res. 58:61–70. 2015. View Article : Google Scholar

217 

Ramos E, Patino P, Reiter RJ, Gil-Martín E, Marco-Contelles J, Parada E, de Los Rios C, Romero A and Egea J: Ischemic brain injury: New insights on the protective role of melatonin. Free Radic Biol Med. 104:32–53. 2017. View Article : Google Scholar : PubMed/NCBI

218 

Paredes SD, Rancan L, Kireev R, González A, Louzao P, González P, Rodríguez-Bobada C, García C, Vara E and Tresguerres JA: Melatonin counteracts at a transcriptional level the inflammatory and apoptotic response secondary to ischemic brain injury induced by middle cerebral artery blockade in aging rats. Biores Open Access. 4:407–416. 2015. View Article : Google Scholar : PubMed/NCBI

219 

Wang X, Figueroa BE, Stavrovskaya IG, Zhang Y, Sirianni AC, Zhu S, Day AL, Kristal BS and Friedlander RM: Methazolamide and melatonin inhibit mitochondrial cytochrome C release and are neuroprotective in experimental models of ischemic injury. Stroke. 40:1877–1885. 2009. View Article : Google Scholar : PubMed/NCBI

220 

Gilani GS, Nimal RW, Mueller R and Mazza G: Effects of source of protein and supplementary extracted isoflavones and anthocyanins on longevity of Stroke-prone Spontaneously Hypertensive (SHRSP) rats. J Toxicol Sci. 34:335–341. 2009. View Article : Google Scholar : PubMed/NCBI

221 

Song F, Zhu Y, Shi Z, Tian J, Deng X, Ren J, Andrews MC, Ni H, Ling W and Yang Y: Plant food anthocyanins inhibit platelet granule secretion in hypercholesterolaemia: Involving the signalling pathway of PI3K-Akt. Thromb Haemost. 112:981–991. 2014. View Article : Google Scholar : PubMed/NCBI

222 

Feng R, Ni HM, Wang SY, Tourkova IL, Shurin MR, Harada H and Yin XM: Cyanidin-3-rutinoside, a natural polyphenol antioxidant, selectively kills leukemic cells by induction of oxidative stress. J Biol Chem. 282:13468–13476. 2007. View Article : Google Scholar : PubMed/NCBI

223 

Dreiseitel A, Schreier P, Oehme A, Locher S, Rogler G, Piberger H, Hajak G and Sand PG: Inhibition of proteasome activity by anthocyanins and anthocyanidins. Biochem Biophys Res Commun. 372:57–61. 2008. View Article : Google Scholar : PubMed/NCBI

224 

Cai Y, Li X, Pan Z, Zhu Y, Tuo J, Meng Q, Dai G, Yang G and Pan Y: Anthocyanin ameliorates hypoxia and ischemia induced inflammation and apoptosis by increasing autophagic flux in SH-SY5Y cells. Eur J Pharmacol. 883:1733602020. View Article : Google Scholar : PubMed/NCBI

225 

Pan Z, Cui M, Dai G, Yuan T, Li Y, Ji T and Pan Y: Protective effect of anthocyanin on neurovascular unit in cerebral ischemia/reperfusion injury in rats. Front Neurosci. 12:9472018. View Article : Google Scholar

226 

Liobikas J, Skemiene K, Trumbeckaite S and Borutaite V: Anthocyanins in cardioprotection: A path through mitochondria. Pharmacol Res. 113:808–815. 2016. View Article : Google Scholar : PubMed/NCBI

227 

Min J, Yu SW, Baek SH, Nair KM, Bae ON, Bhatt A, Kassab M, Nair MG and Majid A: Neuroprotective effect of cyanidin-3-O-glucoside anthocyanin in mice with focal cerebral ischemia. Neurosci Lett. 500:157–161. 2011. View Article : Google Scholar : PubMed/NCBI

228 

Cui HX, Chen JH, Li JW, Cheng FR and Yuan K: Protection of anthocyanin from myrica rubra against cerebral ischemia-reperfusion injury via modulation of the TLR4/NF-κB and NLRP3 pathways. Moleculs. 23:17882018. View Article : Google Scholar

229 

Gutierrez-Vargas JA, Munera A and Cardona-Gomez GP: CDK5 knockdown prevents hippocampal degeneration and cognitive dysfunction produced by cerebral ischemia. J Cereb Blood Flow Metab. 35:1937–1949. 2015. View Article : Google Scholar : PubMed/NCBI

230 

Xue LX, Zhang T, Zhao YW, Geng Z, Chen JJ and Chen H: Efficacy and safety comparison of DL-3-n-butylphthalide and Cerebrolysin: Effects on neurological and behavioral outcomes in acute ischemic stroke. Exp Ther Med. 11:2015–2020. 2016. View Article : Google Scholar : PubMed/NCBI

231 

Fagan SC, Waller JL, Nichols FT, Edwards DJ, Pettigrew LC, Clark WM, Hall CE, Switzer JA, Ergul A and Hess DC: Minocycline to improve neurologic outcome in stroke (MINOS): A dose-finding study. Stroke. 41:2283–2287. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2022
Volume 49 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Zeng W, Zhang Y, Yu Q, Zeng M, Gan J, Zhang W, Jiang X and Li H: Focus on the role of mitochondria in NLRP3 inflammasome activation: A prospective target for the treatment of ischemic stroke (Review). Int J Mol Med 49: 74, 2022
APA
Zhang, X., Zeng, W., Zhang, Y., Yu, Q., Zeng, M., Gan, J. ... Li, H. (2022). Focus on the role of mitochondria in NLRP3 inflammasome activation: A prospective target for the treatment of ischemic stroke (Review). International Journal of Molecular Medicine, 49, 74. https://doi.org/10.3892/ijmm.2022.5130
MLA
Zhang, X., Zeng, W., Zhang, Y., Yu, Q., Zeng, M., Gan, J., Zhang, W., Jiang, X., Li, H."Focus on the role of mitochondria in NLRP3 inflammasome activation: A prospective target for the treatment of ischemic stroke (Review)". International Journal of Molecular Medicine 49.6 (2022): 74.
Chicago
Zhang, X., Zeng, W., Zhang, Y., Yu, Q., Zeng, M., Gan, J., Zhang, W., Jiang, X., Li, H."Focus on the role of mitochondria in NLRP3 inflammasome activation: A prospective target for the treatment of ischemic stroke (Review)". International Journal of Molecular Medicine 49, no. 6 (2022): 74. https://doi.org/10.3892/ijmm.2022.5130