Open Access

Therapeutic strategies targeting the NLRP3‑mediated inflammatory response and pyroptosis in cerebral ischemia/reperfusion injury (Review)

  • Authors:
    • Wan-Li Duan
    • Xue-Jie Wang
    • Ya-Ping Ma
    • Zhi-Mei Sheng
    • Hao Dong
    • Li-Ying Zhang
    • Bao-Gang Zhang
    • Mao-Tao He
  • View Affiliations

  • Published online on: January 22, 2024     https://doi.org/10.3892/mmr.2024.13170
  • Article Number: 46
  • Copyright: © Duan 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

Ischemic stroke poses a major threat to human health. Therefore, the molecular mechanisms of cerebral ischemia/reperfusion injury (CIRI) need to be further clarified, and the associated treatment approaches require exploration. The NOD‑like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome serves an important role in causing CIRI, and its activation exacerbates the underlying injury. Activation of the NLRP3 inflammasome triggers the maturation and production of the inflammatory molecules IL‑1β and IL‑18, as well as gasdermin‑D‑mediated pyroptosis and CIRI damage. Thus, the NLRP3 inflammasome may be a viable target for the treatment of CIRI. In the present review, the mechanisms of the NLRP3 inflammasome in the intense inflammatory response and pyroptosis induced by CIRI are discussed, and the therapeutic strategies that target the NLRP3‑mediated inflammatory response and pyroptosis in CIRI are summarized. At present, certain drugs have already been studied, highlighting future therapeutic perspectives.

Introduction

Stroke is a leading cause of human death and disability, and poses a major threat to humans (1). In total, ~85% of stroke are caused by cerebral ischemia and 15% are caused by cerebral hemorrhage (2). Cerebral ischemia is the result of a lack of blood supply due to occlusion of the cerebral arteries, which results in a lack of glucose and oxygen supply to all brain cells. Therefore, lack of blood in the brain disturbs intracellular homeostasis, which causes inflammation, oxidative damage, excitotoxicity and finally the death of brain cells (3). Thrombolysis to restore blood supply to the brain is currently a viable treatment option for (4). However, rapid reperfusion can lead to further damage to areas of the brain, a condition known as cerebral ischemia/reperfusion injury (CIRI) (5,6). Nevertheless, there are a number of possible mechanisms by which CIRI can occur, including inflammatory response (7), Ca2+ overload (8), overproduction of reactive oxygen species (ROS) (9), neuronal damage caused by glutamate (10) and mitochondria induced-autophagy (11). Of these mechanisms, neuroinflammation serves a key role in CIRI, including via local cytokine upregulation and leukocyte infiltration (12).

Inflammasomes are protein complexes, and potent substances that activate inflammatory mediators, which was first proposed by Martinon et al (13) in 2002. Inflammasomes are part of the innate immune response of the body against pathogen invasion, inflammasomes are activated by cellular infection or stress stimulation and induce the expression, maturation and release of various pro-inflammatory cytokines like IL-18 and IL-1β, thereby triggering a range of inflammatory responses (14,15). Inflammasomes are mainly composed of the nucleotide-binding oligomeric domain-like receptor (NLR) family, which can be divided into three subfamilies: The NLRP, nucleotide-binding oligomerization domains (NODs) and the ice protease-activating factor (IPAF) subfamilies, including NLR family apoptosis inhibitory protein and IPAF (14).

The NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is the most widely studied inflammasomes and contains NLRP3, Pro-caspase-1 and apoptosis-associated spot-like protein (ASC) (1618) (Fig. 1B). NLRP3 consists of an amino-terminal pyrin domain structural domain, a central nucleotide-binding structural domain and an oligomeric structural domain (19) (Fig. 1A). NLRP3 inflammasome assembly is initiated by the interaction of the pyrin structural domain of NLRP3 with the pyrin structural domain of ASC (20). The NLRP3 inflammasome serves a key role in the innate immune system, and activation of the NLRP3 inflammasome mediates the activation of downstream caspase-1 and secretion of the pro-inflammatory cytokines, IL-1β and IL-18, in response to microbial invasion and cellular damage (21). The NLRP3 inflammasome can be activated by different stimuli, including damage-associated molecular patterns (DAMPS) and pathogen-associated molecular patterns (PAMPs) (16). DAMPS are regulated by the pro-inflammatory pathway, such as toll-like receptor (TLR)/NF-κB) signaling pathway, which increases NLRP3 and IL-1β protein expression (22,23) and reduces the activation threshold of NLRP3 through additional post-translational modifications (2426). PAMPs include Ca2+ signaling disruption, mitochondrial dysfunction, ROS production, K+ efflux and lysosomal rupture, promoting assembly of the inflammasome and activating caspase-1, which catalyzes the conversion of pro-IL-1β to active IL-1β (27,28).

As a novel form of programmed cell death, pyroptosis is mainly induced by the gasdermin (GSDM) family (29,30). Of the six members of the GSDM family, five are closely related to pyroptosis and these are GSDMA, GSDMB, GSDMC, GSDMD and GSDME (31). Members of the GSDM family have highly conserved N-terminal and C-terminal domains, and the N-terminal domain can form pores in the cell membrane, causing pyroptosis (32). Activation of inflammasomes can mediate the scission of GSDMD by caspase, which results in formation of GSDMD-N-terminal and finally leads to pyroptosis (33). In addition, pyroptosis is also a crucial pathophysiological process in ischemic stroke (34).

NLRP3 inflammasome is involved in CIRI

Activation of the NLRP3 inflammasome can induce CIRI (35). Abulafia et al (36) first demonstrated that the NLRP inflammasome serves a key role in the inflammatory response to ischemic stroke. Furthermore, CIRI causes upregulation of NLRP3 expression (37) and inhibition of the NLRP3 inflammasome might exert a neuroprotective effect to attenuate CIRI following stroke onset (38). In addition, the NLRP3 inflammasome inhibitor, MCC950, attenuates cerebral infarction, edema, hemorrhagic transformation and neurological deficits in mice following CIRI (38). Furthermore, NLRP3 inhibition facilitates diabetes-mediated cognitive impairment and vascular neural remodeling after CIRI (39,40). Additionally, the NLRP3 inflammasome drives the inflammatory response in CIRI as revealed in a study by Franke et al (35), which demonstrated that NLRP3 mRNA and protein expression was elevated following CIRI, meanwhile other inflammatory vesicles did not change significantly (NLR family CARD domain-containing 4, absent in melanoma 2 and NLRP1). A disintegrin and metalloproteinase 8 (ADAM8) is a transmembrane protein with a number of different functions that serves an important role in tumor and neuroinflammation-related diseases (41). In addition, there is evidence indicating that ADAM8 can aggravate CIRI by activating the NLRP3 inflammasome (41). Furthermore, the NLRP3 inflammasome is a key causative factor in stroke-induced blood-brain barrier disruption, in which the NLRP3 inflammasome exacerbates CIRI by activating inflammatory signaling cascades, inducing pyroptosis of brain endothelial cells and promoting disruption of the blood-brain barrier (42). Early inhibition or blockade of NLRP3 activation protects against CIRI by reducing inflammation and stabilizing the blood-brain barrier (42). In summary, the NLRP3 inflammasome activation is one of the key mechanisms for CIRI. Therefore, further research that focuses on the NLRP3 inflammasome as a therapeutic target for CIRI and the prevention and treatment of ischemic stroke is essential.

Mechanisms of the NLRP3 inflammasome in CIRI

ROS-mediated activation of the NLRP3 inflammasome

Oxidative stress is known to be implicated in the pathogenesis of CIRI, and a study has demonstrated that oxidative stress serves an important role in the prevention and treatment of ischemic stroke by regulating the level of inflammation (43). Oxidative stress can produce ROS. ROS are radicals containing oxygen atoms, and include H2O2, O2− and OH. ROS are mainly derived from the mitochondria and can also be produced by cellular enzymes, including lipoxygenase and cyclooxygenase, which are responsible for inflammasome activation (44). CIRI takes place when the tissue damage caused by restoration of the blood supply to the tissue after a period of ischemia causes tissue damage. This reconstitution of blood flow causes accumulation of ROS, disturbance of cellular ion homeostasis and induce inflammatory response, thereby triggering further damage to ischemic tissues (44). In particular, ROS induce NLRP3 inflammasome activation and stimulate tissue inflammation during CIRI (44,45). Furthermore, ROS have been demonstrated to be a proximal signal for NLRP3 inflammasome activation in inflammatory diseases including CIRI, renal and cardiac ischemia-reperfusion (46). Pro-oxidant and pro-inflammatory thioredoxin-interacting protein (TXNIP), a key regulator of ROS, is associated with inflammation (47). TXNIP is required for NLRP3 activation, which leads to the initiation or worsening of the disease state (48). The increase in ROS generation leads to the upregulation of thioredoxin, TXNIP recruitment of NLRP3 and NLRP3 activation (49). TXNIP is activated by ROS and promotes NLRP3 inflammasome activation by binding to NLRP3 following ischemic stroke (Fig. 2), and inhibition of TXNIP expression reduces inflammasome activation after ischemic stroke (49,50). Mitochondria also serve an important role in the regulation of ROS. Uncoupling protein 2 (UCP2) is an inner membrane protein of the mitochondria that has been reported to regulate mitochondrial potential and ROS production (51,52). At present, there is a study has reported that UCP2 serves an important role in CIRI. UCP2 deficiency aggravates hyperglycemia-induced CIRI by enhancing NLRP3 inflammasome activation and ROS generation (53). Since ROS are an important activator of NLRP3 following CIRI, strategies that eliminate excessive ROS may be effective therapeutic approaches for ischemic stroke.

Activation of the TLR4/NF-κB signaling pathway mediates upregulation of NLRP3 inflammasome expression

TLR4 is a transmembrane receptor protein of the innate immune system that is upregulated following CIRI (54). Upregulation of TLR4 activates NF-κB, which induces the release of number of proinflammatory factors such as IL-18 and IL-1β, triggering an inflammatory response and leading to brain injury (54). Microglia are intrinsic myeloid cells of the central nervous system and are involved in the development of CIRI. For macrophages or microglia, the presence of an NLRP3 activator alone is not sufficient to induce inflammasome activation, and its activation requires initiation signals (55). NLRP3 inflammasome activation must first be induced by initiating stimuli, such as ligands for TLRs, NLRs (such as NOD1 and NOD2) or cytokine receptors, which activate the transcription factor NF-κB and upregulate NLRP3 and IL-1β expression (55). Previous studies have demonstrated that activation of the TLR4/NF-κB signaling pathway is a fundamental step in the formation of the NLRP3 inflammasome and is closely associated with activation of the NLRP3 inflammasome (56,57). TLR4 serves an important role in CIRI and is widely expressed in the brain, especially in microglia and endothelial cells (58,59). Furthermore, inhibition of the TLR4/NF-κB signaling pathway can reduce CIRI by regulating the inflammatory response and apoptosis (60). Collectively, the aforementioned studies have demonstrated that TLR4 activation is a key factor in the upregulation of NLRP3 expression following CIRI, implying that targeting TLR4 or its downstream proteins is likely to be an effective treatment for ischemic stroke (Fig. 2).

Autophagy can inhibit NLRP3 inflammasome activation

Autophagy acts as a stable self-sustaining process in numerous physiological and pathological processes of eukaryotic cells. In this process, bilayers encapsulate pathogens, abnormal proteins and organelles to form autophagosomes, which are transferred to lysosomes for degradation (61). Autophagy can be classified as macroautophagy, microautophagy and chaperone-mediated autophagy depending on the duration of action, the inducing signal, the type of target and the transit pathway into the lysosome (16,62). Macroautophagy involves the formation of double-membrane vesicles that separate the cytoplasm. These intact vesicles, termed autophagic vesicles, then fuse with lysosomes for subsequent degradation (63,64). In microautophagy, the material to be degraded reaches the lysosomal lumen via lysosomal invagination or the endoplasmic membrane (65,66). Chaperone-mediated autophagy only occurs in mammalian cells and allows for the selective degradation of proteins with specific amino acid sequences (67). Among these three autophagic processes, macroautophagy, commonly termed autophagy, is the most active form and has been extensively studied in disease (68,69). Conserved proteins such as Beclin1, LC3 and P62 are involved in the autophagic process and are considered autophagy-related proteins (63). Autophagy is affected by various parameters such as endoplasmic reticulum stress (ERS), ROS, nutritional deficiencies, immune or inflammatory stimuli, accumulation of organelle damage, and the Ca2+ concentration (70,71). Under physiological conditions, autophagy is typically maintained at basal levels. However, in pathological states, upregulated autophagy removes dysfunctional proteins from cells and aids cell survival (72). Autophagy can inhibit NLRP3 activation by reducing ASC expression, increasing phosphorylation of NLRP3 and scavenging ROS (16). The cytoplasmic protein, activating transcription factor 4 (ATF4), serves an important role in the regulation of autophagy, and ATF4 is a member of the activating transcription factor/cAMP response element binding protein family (73). As a transcription factor, ATF4 was involved in Endoplasmic reticulum (ER) homeostasis, autophagy and inflammation response (73). In addition, ATF4 inhibits the NLRP3 inflammasome-mediated inflammatory response via upregulation of Parkin-dependent mitochondrial autophagy in CIRI (74). Finally, autophagy can target the degradation of IL-1β, inhibit activation of the NLRP3 inflammasome and reduce the release of inflammatory cytokines (75,76). Thus, autophagy has been shown to negatively regulate the NLRP3 inflammasome activation and effectively reduce CIRI (Fig. 2).

Other pathways mediating the inhibition of the NLRP3 inflammasome

In addition to the aforementioned three methods of activation, NLRP3 can also be activated by other pathways following CIRI. For example, there is evidence that the α7 nicotinic acetylcholine receptor (α7nAChR) is critical in mediating cholinergic anti-inflammatory signaling (77). Electroacupuncture promotes α7nAChR-mediated inhibition of the NLRP3 inflammasome, thereby reducing CIRI and neuroinflammation (78), which implies that α7nAChR may be an upstream signal for NLRP3 activation. ERS is severe in ischemic brain injury and leads to an inflammatory response via activation of caspase-12 (79). In a previous study, pretreatment with the caspase-12 specific inhibitor Z-ATAD-FMK attenuated cell injury and apoptosis, and reduced the levels of NLRP3, caspase-1, IL-1β and cleaved caspase-3 compared with oxygen-glucose deprivation/recovery (OGD/R) group (79). Therefore, the NLRP3 inflammasome signaling pathway may be inhibited by suppression of caspase-12 signaling to attenuate CIRI. In addition, electroacupuncture induces upregulation of neuronal cylindromatosis (CYLD) expression, which exerts anti-inflammatory and neuroprotective effects by inhibiting NLRP3 expression, regulates the interaction between neurons and microglia, reduces M1 microglia in the peri-ischemic cortex, and improves the activation of M2 microglia, thereby reducing CIRI (80). Collectively, the above studies demonstrated that both α7nAChR and CYLD can inhibit NLRP3 inflammasome activation, while ERS-mediated caspase-12 activation can upregulate NLRP3 expression (Fig. 2).

Activation of the NLRP3 inflammasome promotes the release of downstream inflammatory factors and facilitates pyroptosis in CIRI. Activation of inflammasomes has been associated with various inflammatory diseases, including post-ischemic inflammation following ischemic stroke (12). Inflammasomes mediate the activation of caspase-1, which in turn induces the secretion of pro-inflammatory cytokines and pyroptosis (81). Caspase-1 is activated upon recruitment to the inflammasome, then activated caspase-1 cleaves the cytokines Pro-IL-1β and Pro-IL-18 into their mature bioactive forms (13,82). IL-1β controls fever, pain threshold, vasodilation, and hypotension, and promotes immune cell infiltration into infected or damaged tissues (83). IL-18 is required for production of IFN-γ, a costimulatory cytokine that mediates adaptive immunity (84). CIRI activates NLRP3, induces the release of IL-1β and IL-18 and promotes maturation of GSDMD-N, and leads to severe neuronal pyroptosis (85). Previous studies have demonstrated that the expression levels of GSDMD-N, NLRP1/3, IL-1β and IL-18 in Sprague-Dawley rats and mice were increased following CIRI compared with the Sham group, and intervention treatment of these inflammatory factors attenuated CIRI (8690). In another study, the mRNA expression levels of NLRP3, caspase-1, IL-1β, IL-6 and TNF-α were increased in microglia after OGD/R treatment compared with the control group (91). Overall, NLRP3 inflammasome activation promotes the release of downstream inflammatory factors and causes GSDMD-mediated pyroptosis following CIRI (Fig. 2).

Therapeutic strategies targeting NLRP3 in CIRI

Therapeutic strategies that alleviate CIRI by reducing the activation of NLRP3 via ROS inhibition

During CIRI, ROS stimulate tissue inflammation and activate the NLRP3 inflammasome. Inflammatory diseases are often characterized by the activation of the NLRP3 inflammasome, which is primarily triggered by ROS. Therefore, inhibiting the production of ROS or increasing their consumption following CIRI could be a viable treatment option for stroke (92). In a study by Cao et al (93), it was demonstrated that ruscogenin reduced ROS levels following CIRI, which in turn inhibited TXNIP/NLRP3 inflammasome activation and mitigated ischemia-induced blood-brain barrier dysfunction. Additionally, astilbin has been reported to reduce the brain infarct volume and alleviate neurological deficits in middle cerebral artery occlusion (MCAO) rats (94). Furthermore, astilbin has been demonstrated to inhibit cellular inflammation induced by OGD/R by suppressing the activation of the ROS-NLRP3 inflammasome axis (94). Cepharanthine has also been demonstrated to reduce CIRI by inhibiting the 12/15-lipoxygenase signaling pathway, leading to a decrease in ROS and the downregulation of NLRP3 expression (95). In addition, ATN-161 has been indicated to exert a protective effect on cells by reducing the levels of mitochondrial superoxide radicals, thereby alleviating oxidative stress and intracellular ROS during the onset of CIRI (96). However, tomentosin promotes the production of superoxide dismutase in rats during CIRI, which scavenges free radicals, accelerates the antioxidant system, inhibits NLRP3 signaling and attenuates CIRI (97). Oleanolic acid (OA) has been demonstrated to reduce microglia activation and ROS in CIRI, suggesting that OA may exert neuroprotective effects on ischemic stroke by inhibiting NLRP3 inflammasome activation through the reduction of ROS (98). The aforementioned studies have demonstrated that decreasing ROS levels can mitigate the harm caused by CIRI or cellular OGD/R treatment. Therefore, inhibiting the ROS may be a viable option for the treatment of stroke (Table I).

Table I.

Therapeutic strategies using the NLRP3 inflammasome as a target in cerebral ischemia/reperfusion.

Table I.

Therapeutic strategies using the NLRP3 inflammasome as a target in cerebral ischemia/reperfusion.

First author/s, yearDrug or treatmentManner of NLRP3 inhibitionMode of action(Refs.)
Cao G, 2016RuscogeninReduces ROSReduces ROS levels(93)
Li Y, 2020Astilbin Inhibits ROS-NLRP3 axis(94)
Zhao J, 2020Cepharanthine Inhibits 12/15-lox signaling pathway(95)
Amruta N, 2021ATN-161 Reduces mtROS and intracellular ROS(96)
He J, 2020Tomentosin Enhances antioxidant system(97)
Sapkota A, 2022Oleanolic acid Attenuates ROS(98)
Cui HX, 2018PAESInhibits TLR4/NF-κB signaling pathwayInhibits the TLR4/NF-κB/NLRP3 signaling pathway(99)
Ye Y, 2019Meisoindigo Inhibits the TLR4/NF-κB signaling pathway(100)
Dai M, 2020D-carvone Inhibits the TLR4 signaling pathway(101)
Wang K, 2020Exosomes treated with melatonin Modulates the TLR4/NF-κB signaling pathway(102)
He J, 2020Tomentosin Reduces TLR4 expression(97)
Han D, 2020Vinpocetine Inhibits the NF-κB signaling pathway(103)
Liu J, 2021Salidroside Inhibits TLR4/NF-κB signaling pathway(104)
Ran Y, 2021Curcumin Inhibits the NF-κB/NLRP3 signaling pathway(105)
Zeng et al, 2020BMSC-ExosEnhances autophagy-mediated inhibition ofPromotes the AMPK/mTOR signaling pathway(107)
NLRP3 activation
Hu Z, 2021MSC-exos Upregulates FOXO3a expression(108)
Huang Z, 2021Pien-Tze-Huang Promotes the AMPK/mTOR/ULK1 signaling pathway(109)
Ma C, 2019QingkailingOther pathwayInhibit AMPK-mediated NLRP3 activation(110)
Xiao L, 2021AST IV Activates Nrf2(111)
Jiang T, 2019 & Lin X, 2021 Electroacupuncture Promotes α7nAChR and CYLD-mediated inhibition of NLRP3(78,80)
Tang H, 2022VNS Promotes inhibition of NLRP3 by α7nAChR(112)
Liu L, 2020Z-ATAD-FMK Inhibits caspase-12(79)
Zhang Y, 2021GLGZG Activates the PI3K/AKT signaling pathway(113)
Wang B, 2021Tongxinluo Reduces NLRP3 expression(114)
Mo ZT, 2021ICA Inhibits the IRE1/XBP1s signaling pathway(91)
Shi M, 2022Remimazolam Downregulates NLRP3 expression(89)
Zhu et al, 2022Xingxiong injection Activates the AKT/Nrf2 signaling pathway(90)

[i] α7nAChR, α7 nicotinic acetylcholine receptor; 12/15-lox, 12/15-lipoxygenase; AMPK, AMP-activated protein kinase; AST IV, astragaloside IV; BMSC-Exos, exosomes secreted from bone marrow mesenchymal stem cells; CYLD, cylindromatosis; GLGZG, Gualou Guizhi granule; ICA, icariin; IRE1, inositol-requiring enzyme type 1; MSC-exos, mesenchymal stem cell-derived exosomes; NLRP3, NOD-like receptor thermal protein domain associated protein 3; Nrf2, nuclear factor erythroid 2-related factor 2; PAES, purified anthocyanin extracts; ROS, reactive oxygen species; TLR4, toll-like receptor 4; VNS, vagus nerve stimulation; XBP1s, spliced form of X-box binding protein 1; mtROS, mitochondrial reactive oxygen species; ULK1, UNC-51-like kinase 1.

Therapeutic strategies that attenuate CIRI by inhibiting TRL4-mediated NLRP3 upregulation

TLR4 is an important factor in CIRI, and its downstream NF-κB signaling pathway is crucial in the formation of the NLRP3 inflammasome and is closely linked to its activation (54). Inhibiting the TLR4/NF-κB signaling pathway at the onset of CIRI may be a viable treatment option for stroke (54). Cui et al (99) conducted a study on anthocyanin derived from Myrica rubra, and revealed that treatment of ischemia/reperfusion (I/R) mice with purified anthocyanin extracts for 1 week resulted in a decrease in brain infarct volume, disease damage, and nitric oxide and malondialdehyde levels, while superoxide dismutase levels were increased compared with Sham group (99). In addition, treatment with meisoindigo resulted in improvements in neurological scores, reduced infarct volume and decreased brain edema in MCAO/R mice compared with Sham group. Further analysis revealed that meisoindigo inhibited the expression of TLR4/NF-κB signaling pathway-related proteins in a dose-dependent manner. This inhibition led to the downregulation of NLRP3, high mobility group box 1 and IL-1β expression (100). D-carvone has been reported to inhibit the TLR4-induced signaling pathway of inflammatory cytokines and reduce NLRP3 expression, leading to the successful amelioration of I/R-induced neuroinflammation in the brains of rats. As a result, I/R-induced brain injury in the hippocampal and cortical regions was attenuated (101). Exosomes treated with melatonin have been shown to effectively reduce the infarct size and improve functional recovery by modulating the TLR4/NF-κB signaling pathway and reducing NLRP3-induced inflammation following CIRI (102). Additionally, tomentosin treatment enhances antioxidant capacity to reduce ROS levels, while also reducing the expression of TLR4 and its downstream pro-inflammatory cytokines. This ultimately inhibits NLRP3 expression and attenuates CIRI (97). Vinpocetine has been revealed to inhibit the NF-κB pathway-related proteins, which in turn downregulates NLRP3 expression levels. This inhibition leads to a reduction in the release of pro-inflammatory cytokines, resulting in a decrease in the size of cerebral infarcts and an improvement in behavioral recovery in MCAO mice (103). Salidroside has been demonstrated to reverse NLRP3 inflammasome activation, resulting in downregulated levels of NLRP3, ASC, caspase-1, IL-1β and IL-18 proteins, as well as the suppression of key components of the TLR4 signaling pathway in BV2 cells following OGD/R (104). The specific TLR4 inhibitor, TAK242, exhibited the same effect as salidroside on BV2 cells following OGD/R induction, indicating that salidroside has the capability to specifically inhibit the TLR4/NF-κB signaling pathway, reducing NLRP3 expression and attenuating CIRI (104). Curcumin has been demonstrated to attenuate white matter damage caused by stroke to some extent by inhibiting the NF-κB/NLRP3 signaling pathway, improving functional outcomes and reducing microglia apoptosis (105). In summary, the aforementioned studies demonstrated that inhibiting the TLR4/NF-κB signaling pathway through pharmacological treatment can effectively suppress the expression and activation of NLRP3, thereby reducing the inflammatory response and cellular damage caused by CIRI. Furthermore, inhibiting the upregulation of NLRP3 expression mediated by TLR4 may be a viable clinical treatment option for stroke (Table I).

Therapeutic strategies that mitigate CIRI by enhancing the autophagy-mediated inhibition of NLRP3 activation

Autophagy serves a crucial role in various pathophysiological processes such as renal and cardiac ischemia-reperfusion and CIRI. In pathological conditions, autophagy can hinder the activation of the NLRP3 inflammasome by eliminating endogenous inflammasome activators such as ROS, cytokines and damaged mitochondria from inflammatory components. Inducing cellular autophagy through pharmacological intervention during the onset of CIRI may be a viable option for treating patients following ischemic stroke (106). Exosomes secreted from bone marrow mesenchymal stem cells (BMSC-Exos) have been found to increase autophagic flux in PC12 cells treated with OGD/R, while also inhibiting OGD/R-induced pyroptosis (107). Experimental data further indicated that BMSC-Exos treatment led to decreased NLRP3 expression, as well as elevated LC3 II/I and phosphorylated-AMPK)/AMPK levels (107). These findings suggested that BMSC-Exos promoted autophagic flux in PC12 cells via the AMPK/mTOR signaling pathway, while also inhibiting NLRP3 inflammasome-mediated pyroptosis (107). As a result, BMSC-Exos offer protective benefits to PC12 cells, shielding the cells from OGD/R injury (107). In a similar study, it was identified that human umbilical cord mesenchymal stem cell-derived exosomes (MSC-Exos) had a positive impact on BV2 cell viability following OGD/R (108). Additionally, the expression levels of NLRP3, cleaved caspase-1 and GSDMD-N, as well as the release of IL-1β and IL-18, were decreased, while translocase of outer mitochondrial membrane 20 and cytochrome c oxidase subunit 4 isoform 1 expression was increased. However, the neuroprotective effect of MSC-Exos was partially abolished by FOXO3a small interfering RNA treatment, which also attenuated the inhibition of mitochondrial phagocytosis and pyroptosis induced by MSC-Exos treatment. This study suggests that FOXO3a expression is increased by MSC-exos, which in turn enhances mitochondrial autophagy in microglia. MSC-Exos treatment inhibits pyroptosis induced by CIRI and ultimately reduces nerve damage (108). Pien-Tze-Huang has been demonstrated to regulate essential autophagic proteins via the AMPK/mTOR/unc-51 like autophagy activating kinase 1 (ULK1)-related signaling pathway. This regulation enhances the autophagic response and inhibits the production of key pro-inflammatory mediators, as well as the expression of NLRP3 and caspase-1 p20 proteins in lipopolysaccharide-induced BV2 cells. These findings suggest that Pien-Tze-Huang may enhance autophagy following CIRI via the AMPK/mTOR/ULK1 signaling pathway, thereby reducing NLRP3-associated neuroinflammation (109). In summary, the aforementioned studies all indicated that activation of the NLRP3 inflammasome can be effectively suppressed by enhancing autophagy-mediated inhibition of NLRP3 activation. This in turn can lead to a protection in CIRI (Table I).

Therapeutic strategies that attenuate CIRI by inhibiting NLRP3 activation or expression through other pathways

In addition to inhibiting the activation of the NLRP3 inflammasome by reducing ROS, regulating the TLR4/NF-κB pathway or enhancing autophagy, there are a number of other therapeutic strategies available to inhibit NLRP3 through different signaling pathways (Table I). The NLRP3 inflammasome serves a crucial role in regulating the release of inflammatory factors and GSDMD-mediated pyroptosis in CIRI. Inhibiting NLRP3 activation or expression can effectively reduce the injury caused by CIRI (18). It has been demonstrated that Qingkailing can effectively reduce the inflammatory response following CIRI, which is achieved by inhibiting AMPK-mediated NLRP3 activation and in turn attenuating CIRI (110). Similarly, astragaloside IV has been demonstrated to alleviate CIRI by inhibiting NLRP3 inflammasome-mediated apoptosis through the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) (111). Additionally, electroacupuncture has been demonstrated to promote α7nAChR and CYLD-mediated inhibition of the NLRP3 inflammasome, thereby reducing CIRI and neuroinflammation (78,80). The use of vagus nerve stimulation (VNS) treatment has been found to inhibit expression of pyroptosis-related molecules, as well as reduce the number of pyrogenic cells and membrane pores. Notably, α7nAChR agonists have been found to mimic the neuroprotective effects of VNS, which suggests that VNS serves a protective role in CIRI by promoting α7nAChR inhibition of NLRP3-mediated pyroptosis (112). The caspase-12-specific inhibitor, Z-ATAD-FMK, has been reported to reduce cell injury and apoptosis in an OGD/R treatment group by inhibiting the activation of NLRP3. This inhibition also resulted in decreased levels of caspase-1, IL-1β and cleaved caspase-3 compared with control group, indicating that CIRI could be alleviated by inhibiting caspase-12 (79). Gualou Guizhi granule (GLGZG) has been found to effectively reduce CIRI by activating the PI3K/AKT signaling pathway and inhibiting cellular pyroptosis. Additionally, GLGZG suppresses NLRP3 expression and the release of its downstream inflammatory factors (113). Another study found that Tongxinluo can inhibit the pyroptosis of astrocytes during the onset of CIRI. Furthermore, Tongxinluo reduces the expression of NLRP3, caspase-11/1, IL-1β and IL6, and attenuates CIRI by decreasing the accumulation of amyloid-β peptide (114). Icariin has been demonstrated to reduce NLRP3 expression by inhibiting the inositol-requiring enzyme 1/X-box binding protein 1 signaling pathway, which decreases the expression of downstream inflammatory factors, reducing pyroptosis and attenuating CIRI (91). In addition, remimazolam has been reported to downregulate the expression of NLRP3 and its associated released inflammatory factors IL-18 and IL-1β, as well as GSDMD, in MCAO rats. This suggests that remimazolam may serve a protective role against CIRI by inhibiting NLRP3 (89). Similarly, Xingxiong injection administration has been demonstrated to activate the AKT/Nrf2 signaling pathway and inhibit the NLRP3 inflammasome during the onset of CIRI, thereby exerting a protective effect (90).

Conclusion

Activation of the NLRP3 inflammasome is critical for the mechanisms of CIRI. In the present review, the mechanisms of NLRP3 activation during the onset of CIRI are discussed and are shown in Fig. 2. ROS and TLR4 can promote activation of the NLRP3 inflammasome and its downstream inflammatory response. To some extent, autophagy can negatively regulate NLRP3 activation, which has protected CIRI. Additionally, α7nAChR and CYLD activation can inhibit NLRP3, while caspase-12 activates the NLRP3 inflammasome. Activation of NLRP3 ultimately leads to an inflammatory response, as well as GSDMD-mediated pyroptosis. Furthermore, in the studies described previously have demonstrated that specifically inhibiting the NLRP3 inflammasome can mitigate neuroinflammation and improve outcomes following CIRI. The present review also examines current therapeutic approaches that aim to inhibit the NLRP3 inflammasome to reduce the inflammatory response and pyroptosis during the onset of CIRI (Table I). As such, the present review offers a thorough theoretical foundation for conducting fundamental research on CIRI. Specifically, it provides a detailed overview of the mechanism of action of the NLRP3 inflammasome during CIRI, which will serve as a basis for future research in this field. It is recommended that further research also investigates the role of the NLRP3 inflammasome in pathogenesis and identifies novel therapies. The NLRP3 inflammasome may be considered a crucial target for the treatment of CIRI and may broaden the therapeutic field of ischemic stroke.

To the best of our knowledge, the present review was the first to categorize drugs that serve a protective role in CIRI by targeting the NLRP3 inflammasome with different molecular mechanisms. This provides novel strategies for the clinical treatment of ischemic stroke as well as novel ideas for other diseases in which the NLRP3 inflammasome serves a critical role in the pathologic process. For example, Pien-Tze-Huang is an herbal medicine used for a variety of inflammatory diseases, whether Pien-Tze-Huang has a protective effect in hemorrhagic stroke or in renal ischemic reperfusion is also a question that deserves in-depth exploration. Exploring whether drugs that are protective in CIRI by targeting NLRP3 inflammasome also exert protective roles in other inflammatory diseases will contribute to the greater social value and economic benefits.

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 82101410) and the Medicine and Health Science and Technology Development Plan Project of Shandong (grant no. 202101040805).

Availability of data and materials

Not applicable.

Authors' contributions

WLD, XJW and MTH conceived the study. WLD, YPM, ZMS and HD were involved in literature search, data collection and writing. LYZ, BGZ and MTH reviewed and edited the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the 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.

Glossary

Abbreviations

Abbreviations:

CIRI

cerebral ischemia/reperfusion injury

NLRP3

NOD-like receptor thermal protein domain associated protein 3

GSDMD

gasdermin-D

ROS

reactive oxygen species

NLR

nucleotide-binding oligomeric domain-like receptor

ASC

apoptosis-associated spot-like protein

DAMPs

damage-associated molecular patterns

PAMPs

pathogen-associated molecular patterns

TLR

toll-like receptor

ADAM8

a disintegrin and metalloproteinase 8

TXNIP

thioredoxin-interacting protein

UCP2

uncoupling protein 2

ERS

endoplasmic reticulum stress

α7nAChR

α7 nicotinic acetylcholine receptor

OGD/R

oxygen-glucose deprivation/recovery

CYLD

cylindromatosis

OA

oleanolic acid

BMSC-Exos

exosomes secreted from bone marrow mesenchymal stem cells

MSC-exos

mesenchymal stem cell-derived exosomes

VNS

vagus nerve stimulation

GLGZG

Gualou Guizhi granule

ICA

icariin

References

1 

Saini V, Guada L and Yavagal DR: Global epidemiology of stroke and access to acute ischemic stroke interventions. Neurology. 97 (20 Suppl 2):S6–S16. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S, Chiuve SE, Cushman M, Delling FN, Deo R, et al: Heart disease and stroke statistics-2018 update: A report from the American heart association. Circulation. 137:e67–e492. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Yang JL, Mukda S and Chen SD: Diverse roles of mitochondria in ischemic stroke. Redox Biol. 16:263–275. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Wu D, Chen J, Wu L, Lee H, Shi J, Zhang M, Ma Y, He X, Zhu Z, Yan F, et al: A clinically relevant model of focal embolic cerebral ischemia by thrombus and thrombolysis in rhesus monkeys. Nat Protoc. 17:2054–2084. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Huang J, Chen L, Yao ZM, Sun XR, Tong XH and Dong SY: The role of mitochondrial dynamics in cerebral ischemia-reperfusion injury. Biomed Pharmacother. 162:1146712023. View Article : Google Scholar : PubMed/NCBI

6 

An H, Zhou B and Ji X: Mitochondrial quality control in acute ischemic stroke. J Cereb Blood Flow Metab. 41:3157–3170. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Monsour M and Borlongan CV: The central role of peripheral inflammation in ischemic stroke. J Cereb Blood Flow Metab. 43:622–641. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Ludhiadch A, Sharma R, Muriki A and Munshi A: Role of calcium homeostasis in ischemic stroke: A review. CNS Neurol Disord Drug Targets. 21:52–61. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Zheng D, Liu J, Piao H, Zhu Z, Wei R and Liu K: ROS-triggered endothelial cell death mechanisms: Focus on pyroptosis, parthanatos, and ferroptosis. Front Immunol. 13:10392412022. View Article : Google Scholar : PubMed/NCBI

10 

Chamorro Á, Dirnagl U, Urra X and Planas AM: Neuroprotection in acute stroke: Targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol. 15:869–881. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Lugovaya AV, Emanuel TS, Kalinina NM, Mitreikin VP, Artemova AV and Makienko AA: The role of autophagy in the regulation of neuroinflammation in acute ischemic stroke (review of literature). Klin Lab Diagn. 67:391–398. 2022.PubMed/NCBI

12 

Jurcau A and Simion A: Neuroinflammation in cerebral ischemia and ischemia/reperfusion injuries: From pathophysiology to therapeutic strategies. Int J Mol Sci. 23:142021. View Article : Google Scholar : PubMed/NCBI

13 

Martinon F, Burns K and Tschopp J: The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 10:417–426. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Jia S, Yang H, Huang F and Fan W: Systemic inflammation, neuroinflammation and perioperative neurocognitive disorders. Inflamm Res. 72:1895–1907. 2023. View Article : Google Scholar : PubMed/NCBI

15 

Vringer E and Tait SWG: Mitochondria and cell death-associated inflammation. Cell Death Differ. 30:304–312. 2023. View Article : Google Scholar : PubMed/NCBI

16 

Zhao S, Li X, Wang J and Wang H: The role of the effects of autophagy on NLRP3 inflammasome in inflammatory nervous system diseases. Front Cell Dev Biol. 9:6574782021. View Article : Google Scholar : PubMed/NCBI

17 

Fu J and Wu H: Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu Rev Immunol. 41:301–316. 2023. View Article : Google Scholar : PubMed/NCBI

18 

Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD and Latz E: Targeting the NLRP3 inflammasome in inflammatory diseases. Nat Rev Drug Discov. 17:588–606. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Huang Y, Xu W and Zhou R: NLRP3 inflammasome activation and cell death. Cell Mol Immunol. 18:2114–2127. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Vajjhala PR, Mirams RE and Hill JM: Multiple binding sites on the pyrin domain of ASC protein allow self-association and interaction with NLRP3 protein. J Biol Chem. 287:41732–31743. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Kelley N, Jeltema D, Duan Y and He Y: The NLRP3 inflammasome: An overview of mechanisms of activation and regulation. Int J Mol Sci. 20:33282019. View Article : Google Scholar : PubMed/NCBI

22 

Abais JM, Xia M, Zhang Y, Boini KM and Li PL: Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Signal. 22:1111–1129. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Toldo S and Abbate A: The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 15:203–214. 2018. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Yang Y, Wang H, Kouadir M, Song H and Shi F: Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 10:1282019. View Article : Google Scholar : PubMed/NCBI

26 

Nunes PR, Mattioli SV and Sandrim VC: NLRP3 activation and its relationship to endothelial dysfunction and oxidative stress: Implications for preeclampsia and pharmacological interventions. Cells. 10:28282021. View Article : Google Scholar : PubMed/NCBI

27 

Schroder K and Tschopp J: The inflammasomes. Cell. 140:821–832. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Xu J and Núñez G: The NLRP3 inflammasome: Activation and regulation. Trends Biochem Sci. 48:331–344. 2023. View Article : Google Scholar : PubMed/NCBI

29 

Frank D and Vince JE: Pyroptosis versus necroptosis: Similarities, differences, and crosstalk. Cell Death Differ. 26:99–114. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Shi J, Gao W and Shao F: Pyroptosis: Gasdermin-mediated programmed necrotic cell death. Trends Biochem Sci. 42:245–254. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Wang C and Ruan J: Mechanistic insights into gasdermin pore formation and regulation in pyroptosis. J Mol Biol. 434:1672972022. View Article : Google Scholar : PubMed/NCBI

32 

Zou J, Zheng Y, Huang Y, Tang D, Kang R and Chen R: The versatile gasdermin family: Their function and roles in diseases. Front Immunol. 12:7515332021. View Article : Google Scholar : PubMed/NCBI

33 

Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H and Lieberman J: Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 535:153–158. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Long J, Sun Y, Liu S, Yang S, Chen C, Zhang Z, Chu S, Yang Y, Pei G, Lin M, et al: Targeting pyroptosis as a preventive and therapeutic approach for stroke. Cell Death Discov. 9:1552023. View Article : Google Scholar : PubMed/NCBI

35 

Franke M, Bieber M, Kraft P, Weber ANR, 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 : PubMed/NCBI

36 

Abulafia DP, de Rivero Vaccari JP, Lozano JD, Lotocki G, Keane RW and Dietrich WD: Inhibition of the inflammasome complex reduces the inflammatory response after thromboembolic stroke in mice. J Cereb Blood Flow Metab. 29:534–544. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Wang L, Ren W, Wu Q, Liu T, Wei Y, Ding J, Zhou C, Xu H and Yang S: NLRP3 inflammasome activation: A therapeutic target for cerebral ischemia-reperfusion injury. Front Mol Neurosci. 15:8474402022. View Article : Google Scholar : PubMed/NCBI

38 

Ismael S, Zhao L, Nasoohi S and Ishrat T: Inhibition of the NLRP3-inflammasome as a potential approach for neuroprotection after stroke. Sci Rep. 8:59712018. View Article : Google Scholar : PubMed/NCBI

39 

Hong P, Li FX, Gu RN, Fang YY, Lai LY, Wang YW, Tao T, Xu SY, You ZJ and Zhang HF: Inhibition of NLRP3 inflammasome ameliorates cerebral ischemia-reperfusion injury in diabetic mice. Neural Plast. 2018:91635212018. View Article : Google Scholar : PubMed/NCBI

40 

Ward R, Li W, Abdul Y, Jackson L, Dong G, Jamil S, Filosa J, Fagan SC and Ergul A: NLRP3 inflammasome inhibition with MCC950 improves diabetes-mediated cognitive impairment and vasoneuronal remodeling after ischemia. Pharmacol Res. 142:237–250. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Lu H, Meng Y, Han X and Zhang W: ADAM8 activates NLRP3 inflammasome to promote cerebral ischemia-reperfusion injury. J Healthc Eng. 2021:30974322021. View Article : Google Scholar : PubMed/NCBI

42 

Bellut M, Papp L, Bieber M, Kraft P, Stoll G and Schuhmann MK: NLPR3 inflammasome inhibition alleviates hypoxic endothelial cell death in vitro and protects blood-brain barrier integrity in murine stroke. Cell Death Dis. 13:202021. View Article : Google Scholar : PubMed/NCBI

43 

Ahmad M, Dar NJ, Bhat ZS, Hussain A, Shah A, Liu H and Graham SH: Inflammation in ischemic stroke: Mechanisms, consequences and possible drug targets. CNS Neurol Disord Drug Targets. 13:1378–1396. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Li P, Li S, Wang L, Li H, Wang Y, Liu H, Wang X, Zhu X, Liu Z, Ye F and Zhang Y: Mitochondrial dysfunction in hearing loss: Oxidative stress, autophagy and NLRP3 inflammasome. Front Cell Dev Biol. 11:11197732023. View Article : Google Scholar : PubMed/NCBI

45 

Minutoli L, Puzzolo D, Rinaldi M, Irrera N, Marini H, Arcoraci V, Bitto A, Crea G, Pisani A, Squadrito F, et al: ROS-mediated NLRP3 inflammasome activation in brain, heart, kidney, and testis ischemia/reperfusion injury. Oxid Med Cell Longev. 2016:21830262016. View Article : Google Scholar : PubMed/NCBI

46 

Abderrazak A, Syrovets T, Couchie D, El Hadri K, Friguet B, Simmet T and Rouis M: NLRP3 inflammasome: From a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biol. 4:296–307. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Mohamed IN, Ishrat T, Fagan SC and El-Remessy AB: Role of inflammasome activation in the pathophysiology of vascular diseases of the neurovascular unit. Antioxid Redox Signal. 22:1188–1206. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Mohamed IN, Li L, Ismael S, Ishrat T and El-Remessy AB: Thioredoxin interacting protein, a key molecular switch between oxidative stress and sterile inflammation in cellular response. World J Diabetes. 12:1979–1999. 2021. View Article : Google Scholar : PubMed/NCBI

49 

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

50 

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 : PubMed/NCBI

51 

Brand MD and Esteves TC: Physiological functions of the mitochondrial uncoupling proteins UCP2 and UCP3. Cell Metab. 2:85–93. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Hass DT and Barnstable CJ: Uncoupling proteins in the mitochondrial defense against oxidative stress. Prog Retin Eye Res. 83:1009412021. View Article : Google Scholar : PubMed/NCBI

53 

Zhang T, He MT, Zhang XP, Jing L and Zhang JZ: Uncoupling protein 2 deficiency enhances NLRP3 inflammasome activation following hyperglycemia-induced exacerbation of cerebral ischemia and reperfusion damage in vitro and in vivo. Neurochem Res. 46:1359–1371. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Huang D, Zhou J, Li W, Zhang L, Wang X and Liu Q: Casticin protected against neuronal injury and inhibited the TLR4/NF-κB pathway after middle cerebral artery occlusion in rats. Pharmacol Res Perspect. 9:e007522021. View Article : Google Scholar : PubMed/NCBI

55 

Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, et al: Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 183:787–791. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Zhong X, Liu M, Yao W, Du K, He M, Jin X, Jiao L, Ma G, Wei B and Wei M: Epigallocatechin-3-gallate attenuates microglial inflammation and neurotoxicity by suppressing the activation of canonical and noncanonical inflammasome via TLR4/NF-κB pathway. Mol Nutr Food Res. 63:e18012302019. View Article : Google Scholar : PubMed/NCBI

57 

Yao L, Cai H, Fang Q, Liu D, Zhan M, Chen L and Du J: Piceatannol alleviates liver ischaemia/reperfusion injury by inhibiting TLR4/NF-κB/NLRP3 in hepatic macrophages. Eur J Pharmacol. 960:1761492023. View Article : Google Scholar : PubMed/NCBI

58 

Zheng Y, Bu J, Yu L, Chen J and Liu H: Nobiletin improves propofol-induced neuroprotection via regulating Akt/mTOR and TLR 4/NF-κB signaling in ischemic brain injury in rats. Biomed Pharmacother. 91:494–503. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Shukla V, Shakya AK, Perez-Pinzon MA and Dave KR: Cerebral ischemic damage in diabetes: An inflammatory perspective. J Neuroinflammation. 14:212017. View Article : Google Scholar : PubMed/NCBI

60 

Wu M, Liu F and Guo Q: Quercetin attenuates hypoxia-ischemia induced brain injury in neonatal rats by inhibiting TLR4/NF-κB signaling pathway. Int Immunopharmacol. 74:1057042019. View Article : Google Scholar : PubMed/NCBI

61 

Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cadwell K, Cecconi F, Choi AMK, et al: Autophagy in major human diseases. EMBO J. 40:e1088632021. View Article : Google Scholar : PubMed/NCBI

62 

Lv S, Wang H and Li X: The role of the interplay between autophagy and NLRP3 inflammasome in metabolic disorders. Front Cell Dev Biol. 9:6341182021. View Article : Google Scholar : PubMed/NCBI

63 

Wang J, Wu D and Wang H: Hydrogen sulfide plays an important protective role by influencing autophagy in diseases. Physiol Res. 68:335–345. 2019.PubMed/NCBI

64 

Zhu Y, Yin Q, Wei D, Yang Z, Du Y and Ma Y: Autophagy in male reproduction. Syst Biol Reprod Med. 65:265–272. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Sahu R, Kaushik S, Clement CC, Cannizzo ES, Scharf B, Follenzi A, Potolicchio I, Nieves E, Cuervo AM and Santambrogio L: Microautophagy of cytosolic proteins by late endosomes. Dev Cell. 20:131–139. 2011. View Article : Google Scholar : PubMed/NCBI

66 

Debnath J, Gammoh N and Ryan KM: Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 24:560–575. 2023. View Article : Google Scholar : PubMed/NCBI

67 

Kaushik S and Cuervo AM: Chaperone-mediated autophagy: A unique way to enter the lysosome world. Trends Cell Biol. 22:407–417. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Ueno T and Komatsu M: Autophagy in the liver: Functions in health and disease. Nat Rev Gastroenterol Hepatol. 14:170–184. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Mizushima N and Komatsu M: Autophagy: Renovation of cells and tissues. Cell. 147:728–741. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Tooze SA and Yoshimori T: The origin of the autophagosomal membrane. Nat Cell Biol. 12:831–835. 2010. View Article : Google Scholar : PubMed/NCBI

71 

Mizushima N, Yoshimori T and Ohsumi Y: The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol. 27:107–132. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Glick D, Barth S and Macleod KF: Autophagy: Cellular and molecular mechanisms. J Pathol. 221:3–12. 2010. View Article : Google Scholar : PubMed/NCBI

73 

McCarty MF: Nutraceutical and dietary strategies for up-regulating macroautophagy. Int J Mol Sci. 23:20542022. View Article : Google Scholar : PubMed/NCBI

74 

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 : PubMed/NCBI

75 

Cao Z, Wang Y, Long Z and He G: Interaction between autophagy and the NLRP3 inflammasome. Acta Biochim Biophys Sin (Shanghai). 51:1087–1095. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Biasizzo M and Kopitar-Jerala N: Interplay between NLRP3 inflammasome and autophagy. Front Immunol. 11:5918032020. View Article : Google Scholar : PubMed/NCBI

77 

Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, et al: Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 421:384–388. 2003. View Article : Google Scholar : PubMed/NCBI

78 

Jiang T, Wu M, Zhang Z, Yan C, Ma Z, He S, Yuan W, Pu K and Wang Q: Electroacupuncture attenuated cerebral ischemic injury and neuroinflammation through α7nAChR-mediated inhibition of NLRP3 inflammasome in stroke rats. Mol Med. 25:222019. View Article : Google Scholar : PubMed/NCBI

79 

Liu L, Chen M, Lin K, Xiang X, Zheng Y and Zhu S: Inhibiting caspase-12 mediated inflammasome activation protects against oxygen-glucose deprivation injury in primary astrocytes. Int J Med Sci. 17:1936–1945. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Lin X, Zhan J, Jiang J and Ren Y: Upregulation of neuronal cylindromatosis expression is essential for electroacupuncture-mediated alleviation of neuroinflammatory injury by regulating microglial polarization in rats subjected to focal cerebral ischemia/reperfusion. J Inflamm Res. 14:2061–2078. 2021. View Article : Google Scholar : PubMed/NCBI

81 

Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A and Morita R: Bruton's tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun. 6:73602015. View Article : Google Scholar : PubMed/NCBI

82 

Franchi L, Warner N, Viani K and Nuñez G: Function of nod-like receptors in microbial recognition and host defense. Immunol Rev. 227:106–128. 2009. View Article : Google Scholar : PubMed/NCBI

83 

Li Y and Jiang Q: Uncoupled pyroptosis and IL-1β secretion downstream of inflammasome signaling. Front Immunol. 14:11283582023. View Article : Google Scholar : PubMed/NCBI

84 

Dinarello CA: Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 27:519–550. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Sun R, Peng M, Xu P, Huang F, Xie Y, Li J, Hong Y, Guo H, Liu Q and Zhu W: Low-density lipoprotein receptor (LDLR) regulates NLRP3-mediated neuronal pyroptosis following cerebral ischemia/reperfusion injury. J Neuroinflammation. 17:3302020. View Article : Google Scholar : PubMed/NCBI

86 

Lyu Z, Chan Y, Li Q, Zhang Q, Liu K, Xiang J, Li X, Cai D, Li Y, Wang B and Yu Z: Destructive effects of pyroptosis on homeostasis of neuron survival associated with the dysfunctional BBB-glymphatic system and amyloid-beta accumulation after cerebral ischemia/reperfusion in rats. Neural Plast. 2021:45043632021. View Article : Google Scholar : PubMed/NCBI

87 

Liu J, Zheng J, Xu Y, Cao W, Wang J, Wang B, Zhao L, Zhang X and Liao W: Enriched environment attenuates pyroptosis to improve functional recovery after cerebral ischemia/reperfusion injury. Front Aging Neurosci. 13:7176442021. View Article : Google Scholar : PubMed/NCBI

88 

Pang YQ, Yang J, Jia CM, Zhang R and Pang Q: Hypoxic preconditioning reduces NLRP3 inflammasome expression and protects against cerebral ischemia/reperfusion injury. Neural Regen Res. 17:395–400. 2022. View Article : Google Scholar : PubMed/NCBI

89 

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

90 

Zhu T, Fang BY, Meng XB, Zhang SX, Wang H, Gao G, Liu F, Wu Y, Hu J, Sun GB and Sun XB: Folium Ginkgo extract and tetramethylpyrazine sodium chloride injection (Xingxiong injection) protects against focal cerebral ischaemia/reperfusion injury via activating the Akt/Nrf2 pathway and inhibiting NLRP3 inflammasome activation. Pharm Biol. 60:195–205. 2022. View Article : Google Scholar : PubMed/NCBI

91 

Mo ZT, Zheng J and Liao YL: Icariin inhibits the expression of IL-1β, IL-6 and TNF-α induced by OGD/R through the IRE1/XBP1s pathway in microglia. Pharm Biol. 59:1473–1479. 2021. View Article : Google Scholar : PubMed/NCBI

92 

Shang S, Sun F, Zhu Y, Yu J, Yu L, Shao W, Wang Z and Yi X: Sevoflurane preconditioning improves neuroinflammation in cerebral ischemia/reperfusion induced rats through ROS-NLRP3 pathway. Neurosci Lett. 801:1371642023. View Article : Google Scholar : PubMed/NCBI

93 

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 : PubMed/NCBI

94 

Li Y, Wang R, Xue L, Yang Y and Zhi F: Astilbin protects against cerebral ischaemia/reperfusion injury by inhibiting cellular apoptosis and ROS-NLRP3 inflammasome axis activation. Int Immunopharmacol. 84:1065712020. View Article : Google Scholar : PubMed/NCBI

95 

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

96 

Amruta N and Bix G: ATN-161 ameliorates ischemia/reperfusion-induced oxidative stress, fibro-inflammation, mitochondrial damage, and apoptosis-mediated tight junction disruption in bEnd.3 cells. Inflammation. 44:2377–2394. 2021. View Article : Google Scholar : PubMed/NCBI

97 

He J, Wu H, Zhou Y and Zheng C: Tomentosin inhibit cerebral ischemia/reperfusion induced inflammatory response via TLR4/NLRP3 signalling pathway-in vivo and in vitro studies. Biomed Pharmacother. 131:1106972020. View Article : Google Scholar : PubMed/NCBI

98 

Sapkota A and Choi JW: Oleanolic acid provides neuroprotection against ischemic stroke through the inhibition of microglial activation and NLRP3 inflammasome activation. Biomol Ther (Seoul). 30:55–63. 2022. View Article : Google Scholar : PubMed/NCBI

99 

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. Molecules. 23:17882018. View Article : Google Scholar : PubMed/NCBI

100 

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 : PubMed/NCBI

101 

Dai M, Wu L, Yu K, Xu R, Wei Y, Chinnathambi A, Alahmadi TA and Zhou M: D-Carvone inhibit cerebral ischemia/reperfusion induced inflammatory response TLR4/NLRP3 signaling pathway. Biomed Pharmacother. 132:1108702020. View Article : Google Scholar : PubMed/NCBI

102 

Wang K, Ru J, Zhang H, Chen J, Lin X, Lin Z, Wen M, Huang L, Ni H, Zhuge Q and Yang S: Melatonin enhances the therapeutic effect of plasma exosomes against cerebral ischemia-induced pyroptosis through the TLR4/NF-κB pathway. Front Neurosci. 14:8482020. View Article : Google Scholar : PubMed/NCBI

103 

Han D, Wang J, Wen L, Sun M, Liu H and Gao Y: Vinpocetine attenuates ischemic stroke through inhibiting NLRP3 inflammasome expression in mice. J Cardiovasc Pharmacol. 77:208–216. 2020. View Article : Google Scholar : PubMed/NCBI

104 

Liu J, Ma W, Zang CH, Wang GD, Zhang SJ, Wu HJ, Zhu KW, Xiang XL, Li CY, Liu KP, et al: Salidroside inhibits NLRP3 inflammasome activation and apoptosis in microglia induced by cerebral ischemia/reperfusion injury by inhibiting the TLR4/NF-κB signaling pathway. Ann Transl Med. 9:16942021. View Article : Google Scholar : PubMed/NCBI

105 

Ran Y, Su W, Gao F, Ding Z, Yang S, Ye L, Chen X, Tian G, Xi J and Liu Z: Curcumin ameliorates white matter injury after ischemic stroke by inhibiting microglia/macrophage pyroptosis through NF-κB suppression and NLRP3 inflammasome inhibition. Oxid Med Cell Longev. 2021:15521272021. View Article : Google Scholar : PubMed/NCBI

106 

Zhao Y, Hong Z, Lin Y, Shen W, Yang Y, Zuo Z and Hu X: Exercise pretreatment alleviates neuroinflammation and oxidative stress by TFEB-mediated autophagic flux in mice with ischemic stroke. Exp Neurol. 364:1143802023. View Article : Google Scholar : PubMed/NCBI

107 

Zeng Q, Zhou Y, Liang D, He H, Liu X, Zhu R, Zhang M, Luo X, Wang Y and Huang G: Exosomes secreted from bone marrow mesenchymal stem cells attenuate oxygen-glucose deprivation/reoxygenation-induced pyroptosis in PC12 cells by promoting AMPK-dependent autophagic flux. Front Cell Neurosci. 14:1822020. View Article : Google Scholar : PubMed/NCBI

108 

Hu Z, Yuan Y, Zhang X, Lu Y, Dong N, Jiang X, Xu J and Zheng D: Human umbilical cord mesenchymal stem cell-derived exosomes attenuate oxygen-glucose deprivation/reperfusion-induced microglial pyroptosis by promoting FOXO3a-dependent mitophagy. Oxid Med Cell Longev. 2021:62197152021. View Article : Google Scholar : PubMed/NCBI

109 

Huang Z, Zhou X, Zhang X, Huang L, Sun Y, Cheng Z, Xu W, Li CG, Zheng Y and Huang M: Pien-Tze-Huang, a Chinese patent formula, attenuates NLRP3 inflammasome-related neuroinflammation by enhancing autophagy via the AMPK/mTOR/ULK1 signaling pathway. Biomed Pharmacother. 141:1118142021. View Article : Google Scholar : PubMed/NCBI

110 

Ma C, Wang X, Xu T, Yu X, Zhang S, Liu S, Gao Y, Fan S, Li C, Zhai C, et al: Qingkailing injection ameliorates cerebral ischemia-reperfusion injury and modulates the AMPK/NLRP3 inflammasome signalling pathway. BMC Complement Altern Med. 19:3202019. View Article : Google Scholar : PubMed/NCBI

111 

Xiao L, Dai Z, Tang W, Liu C and Tang B: Astragaloside IV alleviates cerebral ischemia-reperfusion injury through NLRP3 inflammasome-mediated pyroptosis inhibition via activating Nrf2. Oxid Med Cell Longev. 2021:99255612021. View Article : Google Scholar : PubMed/NCBI

112 

Tang H, Li J, Zhou Q, Li S, Xie C, Niu L, Ma J and Li C: Vagus nerve stimulation alleviated cerebral ischemia and reperfusion injury in rats by inhibiting pyroptosis via α7 nicotinic acetylcholine receptor. Cell Death Discov. 8:542022. View Article : Google Scholar : PubMed/NCBI

113 

Zhang Y, Wang H, Li H, Nan L, Xu W, Lin Y and Chu K: Gualou guizhi granule protects against OGD/R-induced injury by inhibiting cell pyroptosis via the PI3K/Akt signaling pathway. Evid Based Complement Alternat Med. 2021:66135722021.PubMed/NCBI

114 

Wang B, Lyu Z, Chan Y, Li Q, Zhang L, Liu K, Li Y and Yu Z: Tongxinluo exerts inhibitory effects on pyroptosis and amyloid-β peptide accumulation after cerebral ischemia/reperfusion in rats. Evid Based Complement Alternat Med. 2021:57886022021.PubMed/NCBI

Related Articles

Journal Cover

March-2024
Volume 29 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Duan W, Wang X, Ma Y, Sheng Z, Dong H, Zhang L, Zhang B and He M: Therapeutic strategies targeting the NLRP3‑mediated inflammatory response and pyroptosis in cerebral ischemia/reperfusion injury (Review). Mol Med Rep 29: 46, 2024
APA
Duan, W., Wang, X., Ma, Y., Sheng, Z., Dong, H., Zhang, L. ... He, M. (2024). Therapeutic strategies targeting the NLRP3‑mediated inflammatory response and pyroptosis in cerebral ischemia/reperfusion injury (Review). Molecular Medicine Reports, 29, 46. https://doi.org/10.3892/mmr.2024.13170
MLA
Duan, W., Wang, X., Ma, Y., Sheng, Z., Dong, H., Zhang, L., Zhang, B., He, M."Therapeutic strategies targeting the NLRP3‑mediated inflammatory response and pyroptosis in cerebral ischemia/reperfusion injury (Review)". Molecular Medicine Reports 29.3 (2024): 46.
Chicago
Duan, W., Wang, X., Ma, Y., Sheng, Z., Dong, H., Zhang, L., Zhang, B., He, M."Therapeutic strategies targeting the NLRP3‑mediated inflammatory response and pyroptosis in cerebral ischemia/reperfusion injury (Review)". Molecular Medicine Reports 29, no. 3 (2024): 46. https://doi.org/10.3892/mmr.2024.13170