Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2019 Nov 14;25(12):1299–1307. doi: 10.1111/cns.13262

Update of inflammasome activation in microglia/macrophage in aging and aging‐related disease

Meng‐yan Hu 1, Yin‐yao Lin 1, Bing‐jun Zhang 1, Dan‐li Lu 1, Zheng‐qi Lu 1,, Wei Cai 1,2,
PMCID: PMC6887669  PMID: 31729181

Abstract

Aging and aging‐related CNS diseases are associated with inflammatory status. As an efficient amplifier of immune responses, inflammasome is activated and played detrimental role in aging and aging‐related CNS diseases. Macrophage and microglia display robust inflammasome activation in infectious and sterile inflammation. This review discussed the impact of inflammasome activation in microglia/macrophage on senescence “inflammaging” and aging‐related CNS diseases. The preventive or therapeutic effects of targeting inflammasome on retarding aging process or tackling aging‐related diseases are also discussed.

Keywords: aging, inflammasome, inflammation, macrophage, microglia

1. INTRODUCTION

The aging process of central nervous system (CNS), as well as the pathology of aging‐related diseases, is closely associated with inflammatory responses.1, 2 Inflammasome is a multiprotein complex which is induced in response to microbial invasion or damage‐associated molecular patterns (DAMPs) in innate immune cells.3 Activation of inflammasome results in production of proinflammatory factors, including interleukin (IL)‐1β, and IL‐18, which represents an important amplifier of inflammation. Notably, inflammasome is activated during aging and aging‐related CNS diseases, accelerating the process of senility and CNS disorders at the same time. A broad body of studies have confirmed the key role of microglia and macrophage in aging and aging‐related diseases. Inflammatory milieu during aging‐related CNS diseases activates microglia macrophage, while activation of microglia and macrophage contributes to the exacerbation of neural inflammation in aging‐related disease.4, 5, 6 Microglia and macrophage are the main cells in which inflammasome is potently activated. This review summarizes the impact of inflammasome activation in microglia/macrophage during aging and aging‐related disorders. Preventive or therapeutic effects of targeting inflammasome on tackling aging‐related diseases are also discussed.

2. INFLAMMASOME IS AN AMPLIFIER OF NEURAL INFLAMMATION

Inflammasome is an intracellular complex that detects physiological and pathogenic stimuli. Inflammasome activation was first discovered in myeloid cells, including macrophage/microglia, neutrophil, and dendritic cell.7 Recently, it was demonstrated that other cell types, including, oligodendrocyte, astrocyte, neurons, and epithelial cell, could also trigger inflammasome activation.8, 9, 10, 11 Among inflammasome‐forming cells, it is microglia/macrophage that has the most potent inflammasome activation, thus is most widely studied.12

Classically, inflammasome is composed of sensor, executor, and substrate. Multiple sensors have been found to detect stimuli for inflammasome, including NACHT, LRR, and PYD domains‐containing protein 1 (NLRP1), NLRP2, NLRP3 NLR family CARD domain‐containing protein 4 (NLRC4), and absent in melanoma 2 (AIM2).13 Canonically, sensor of inflammasome recruits the executive enzyme of Caspase‐1 with the adaptor of apoptosis‐associated speck‐like protein, also known as PYCARD (ASC). Subsequently, Caspase‐1 cleaves the substrates of pro‐IL‐1β and pro‐IL‐18 into their active form (Cleaved‐IL1β and Cleaved‐IL18). It is found that Caspase‐8 and Caspase‐11 could also participate in the process of inflammasome activation as executors. Moreover, gasdermin‐D (GSDMD) could be activated by the caspase enzymes (eg, Caspase‐11) and formed pores in cytomembrane of inflammasome‐activating cells, resulting in specific cell death process called pyroptosis.14 Other accessories of inflammasome have been discovered. NIMA‐related kinase 7 (NEK7) has been found to bridge adjacent NLRP3 for their oligomerization and mediate subsequent inflammasome activation.15, 16

The classic understanding of the process of inflammasome formation is based on a two‐signal model (Figure 1). In signal 1, sensors of inflammasome are activated by pathogen‐associated molecular patterns (PAMPs), the signal is passed through by NF‐κB pathway, and transcription of inflammasome‐relevant genes such as NLRP3 and pro‐IL1β is increased.16, 17 In signal 2, DAMPs (including ATP, ROS, Ca2+ mobilization, uric acid, alums, and silica)18 further activate the inflammasome sensors. The sensors then undergo oligomerization and attach to ASC. ASC acts as a molecular platform that recruits pro‐caspase enzymes. The pro‐caspase enzymes are then cleaved into their active form which subsequently cleaves pro‐IL1β and pro‐IL18 into cleaved‐IL1β and cleaved‐IL18. The inflammasome products further exert their inflammatory amplifying effects. In the real battlefield of disease/injury, it is more likely for cells to come across the two kinds of signal concurrently, and the two signals are transmitted at the same time. Moreover, noncanonical activating process of inflammasome is discovered. Lipid A could activate Caspase‐4/5/11 directly, inducing oligomerization of the caspase enzymes, which activates cysteine protease to cleave the downstream substrate of GSDMD.19, 20 Therefore, the two‐signal theory is questioned. Nevertheless, the two‐signal theory of inflammasome activation stills serves as a favorable model for scientific research.

Figure 1.

Figure 1

Two‐signal model of inflammasome signaling. The classic understanding of inflammasome is based on a two‐signal model. In signal 1, sensors (eg, NLRP3) of inflammasome are activated by PAMPs/ DAMPs, leading to activation of NF‐κB pathway and increased transcription of inflammasome‐relevant genes such as NLRP3 and pro‐IL1β. In signal 2, PAMPs/DAMPs (eg, ATP) further activate the inflammasome sensors. The sensors then undergo oligomerization and attach to ASC. ASC acts as a molecular platform that recruits pro‐caspase enzymes. The pro‐caspase enzymes are then cleaved into their active form which subsequently cleave pro‐IL1β and pro‐IL18 into cleaved‐IL1β and cleaved‐IL18

Although the precise mechanism of inflammasome activation is still elusive, there is no doubt that the consequences of inflammasome activation serve as an amplifier of neural inflammation in various aging‐related neurological diseases. Cleaved‐IL1β and cleaved‐IL18 released from inflammasome‐forming cells induce production of proinflammatory factors by neighbor cells, including IFNγ, TNFα, and ROS.21 The proinflammatory factors further impair blood‐brain barrier (BBB) and attract peripheral leukocytes.22, 23 Besides, pyroptosis induced by inflammasome activation gives rise to DAMPs, which activate inflammasome and exacerbate the inflammatory status. Neural inflammation plays a key role in multiple aging‐related neurological diseases. Accumulating evidence reveals that inflammasome activation could aggravate CNS disorders including neurodegenerative diseases and acute CNS stroke. Targeting inflammasome to dampen neural inflammation is a promising therapeutic strategy for the aging‐related neurological disorders.

3. MICROGLIA/MACROPHAGE ARE PRONE TO FORM INFLAMMASOME IN SENESCENT BRAINS IN THE ABSENCE OF DISEASE STATUS

Aging is accompanied by low‐grade inflammation in multiple organs, which is referred as “inflammaging”.24 The persistence of inflammatory status in aged CNS is related to chronic injury and the subsequent functional decline even in the absence of a specific disease. Recently, Furman et al25 reported that the status of inflammaging could be attributed to inflammasome activation. Inflammasome, the efficient inflammation amplifier, was inclined to get activated in aged microglia/macrophage. Multiple components of inflammasome, including NLRC4, Caspse‐1, Caspase‐11, ASC, and IL‐1β, were found to be increased in hippocampus of aged mice.26 In particular, expression of NLRP3, the most studied sensor of inflammasome, was increased in senile microglia compared with their young counterparts.27 Correspondingly, spontaneous activation of Caspase‐1 was detected in aged brain.27 The reason for inclination of inflammasome activation in aged microglia is related to the accumulation of a diverse array of endogenous metabolic danger signals in the microenvironment, including nucleotide metabolites (eg, such as adenine and N4‐acetylcytidine), uric acid, ATP, oxidative stress, cholesterol, lipotoxic fatty acids, and ceramides.27 Some mitochondrial microRNA (eg, let7b, mir‐146a, mir‐133b, mir‐106a, mir‐19b, mir‐20a, mir‐34a, mir‐181a, and mir‐221) and cellular microRNA (eg, miR‐146a, miR‐34a, and miR‐181a) are also involved in inflammasome‐dependent inflammaging.28

3.1. The consequences of inflammasome activation in aged microglia/macrophage are harsh

In aged mice without induction of any diseases, decrease of neurological functions was still evident due to accumulation of daily stress. Ablating NLRP3 inflammasome significantly improved cognitive and motor performance in IL‐1‐dependent manner.27 Recently, Furman et al25 found that the expression of specific inflammasome gene modules stratified the elderly into two extremes: those with high expression of inflammasome‐related genes and those without. The former population was found to have shorter longevity, high rate of hypertension, and arterial stiffness. The phenomenon that aged brains are prone to form inflammasome could be attributed to the development of neurodegenerative diseases and the violent exacerbation of acute neurological disorders in aged individuals. From the inflammasome, inclination in aged brains comes to the enlightenment that inhibiting chronic inflammasome activation might be a potent preventive and curative strategy for aging‐related CNS diseases. Since microglia/macrophage are the major source of inflammasome, targeting the particular cell type should be the most direct and efficient path for inflammasome suppression in aging‐related CNS disorders, in both chronic (eg, neurodegenerative diseases) and acute way (eg, CNS stroke).

4. INFLAMMASOME ACTIVATION IN MICROGLIA/MACROPHAGE IS THE RESULT OF NEURODEGENERATIVE DISEASE DEVELOPMENT AND PROMOTES THE DISEASE PROGRESSION

It is widely accepted that neurodegenerative diseases are associated with chronic CNS inflammation.29, 30 As an efficient inflammation amplifier, inflammasome activation could be the result of neurodegeneration as well as the reason for the disease progression. Specific components in neurodegenerative diseases, such as Amyloid β (Aβ) in Alzheimer's disease (AD) and α‐synuclein in Parkinson's disease (PD), promote inflammasome activation in microglia/macrophage. Meanwhile, activation of inflammasome results in production of highly proinflammatory cytokines such as IL‐1β and IL‐18 by microglia/macrophage, which accelerates disease development.

4.1. Inflammasome activation in Alzheimer's disease

AD causes memory loss and other cognitive impairment in the elderly. In the pathophysiological process of AD, inflammasome formation plays a negligible role. Inflammasome activation is the result of AD development. The extracellular plaque deposition of the Aβ is a principal event in the pathogenesis of AD. Deposition of Aβ causes lysosomal damage and ROS production in microglia, which induces inflammasome activation.31, 32, 33, 34 Moreover, it was reported that Aβ oligomers could directly interact with NLRP3 and ASC in infiltrated macrophages and cause inflammasome activation, which exacerbated neuroinflammation.35, 36 Increase of the inflammasome products (eg, IL‐1β) are detected in serum, cerebrospinal fluid, and brain tissue of AD patients.37 On the other hand, activation of inflammasome promotes AD development. For example, as a product of inflammasome, IL‐1β induces proinflammatory cytokines (eg, IL‐1β, IL‐6 and IL‐18) production and free radical release by glia cells, resulting in neurotoxicity.38 Meanwhile, IL‐1β promotes the production of β‐amyloid precursor protein (APP) and Aβ by neurons, induces phosphorylation of Tau protein, and mediates neurofibrillary tangle formation.33, 39, 40

After successive failure of clinical trial for new AD treatment, research on finding effective drug is imperative. Since inflammasome activation is associated with amyloid genesis and quantitative inflammation mediators, therapeutic approaches targeting inflammasome to postpone AD development have been proposed. Heneka et al41 found that interfering NLRP3 gene expression could reduce Aβ deposition, decrease neurotic plaque burden, and improve memory and behavior functions in AD models. Pharmacologically, suppressing inflammasome activation with ATP inhibitor or purinergic 2X7 (P2X7) receptor antagonist improved age‐related cognitive decline.42 The therapeutic effects of inflammasome suppressive treatments reveal that resisting inflammaging by tackling inflammasome is a promising strategy for AD therapy.

4.2. Inflammasome activation in Parkinson's disease

Parkinson's disease (PD) is a progressive degenerative CNS disorder that affects the motor system. It is characterized by a profound degeneration of dopaminergic neurons in the brain, accompanied by chronic neuroinflammation, mitochondrial dysfunction, and widespread accumulation of α‐synuclein‐rich protein in Lewy's bodies.43, 44, 45 It has been proved that the α‐synuclein is a potent stimulator of inflammasome. Fibrillar α‐synuclein in microglia leads to impairment of mitochondrial endocytosis and lysosomal dysfunction, which cause ROS dysregulation and activate inflammasome.46, 47 Besides, several kinds of microRNA (eg, miR‐7 and miR‐30e), which are increased in PD brains, could directly activate inflammasome in microglia.46, 48 Similar to the situation in AD, inflammasome products released by microglia within substantia nigra trigger the inflammatory cascades in PD brains. Processive inflammasome activation in microglia may be a sustained stimuli of neuroinflammation that drives progressive dopaminergic neuropathology and promotes fibrillar α‐synuclein accumulation.49, 50

Current therapies for PD, including levodopa treatment and deep brain stimulation, can only manage PD symptoms without tackling the pathological alterations in PD brains. The evidence that inflammasome plays a key role in pathophysiological process of PD highlights the potential of repressing inflammasome as a PD therapy.51 It has been proved that suppressing inflammasome activation with a treatment of NLRP3 inhibitor could effectively mitigate motor deficits, nigrostriatal dopaminergic degeneration, and α‐synuclein aggregation in PD model.50, 52 Inflammasome suppression may represent a thorough therapy that deals with the pathophysiology of PD.

5. INFLAMMASOME ACTIVATION INTENSIFIES NEURAL INFLAMMATION AFTER STROKE

Stroke, including ischemic and hemorrhagic subtypes, is one of the leading causes of death worldwide. As an aging‐related acute disorder, nearly three quarters of stroke occur in people over the age of 65.53 With aging, endothelial dysfunction occurs, followed by structural and functional alternation of cerebral microcirculation and microcirculation, increasing the risks for both ischemic and hemorrhagic stroke.54, 55 It has been demonstrated that phagocytic capacity is downregulated in senile microglia/macrophage. When hemorrhagic stroke occurs, the inflammatory milieu further limits phagocytic activities of microglia/macrophage, exacerbating disease severity of hemorrhagic stroke.56 Therefore, aging is an independent risk factor of both ischemic and hemorrhagic stroke and indicates detrimental disease outcomes.57 On the other hand, prognosis of stroke is largely dependent on the intensity of post‐stroke neural inflammation. Inflammasome that activates in CNS‐resident microglia and the infiltrated macrophage plays a decisive role in post‐stroke neural inflammation.

5.1. Inflammasome activation in acute ischemic stroke

Unlike the chronic neurodegenerative diseases, microglia/macrophage in stroke brain are confronted with robust pathological alterations. In ischemic stroke, acute deprivation of blood supply causes rapid necrosis of brain cells within the ischemic lesion. Rupture of necrotic brain cells gives rise to abundant DAMPs. The danger signals then directly activate inflammasome in microglia/macrophage through multiple pathways including NF‐kB and MAPK signaling.58, 59 Dysfunction of organelles during ischemic injury also contributes to inflammasome activation in microglia/macrophage. Dysfunction of the electron transport chain and accumulation of Ca2+ in the mitochondria result in robust ROS release. Stagnated ATP production then causes impairment of Na+/K+‐ATPase pumps which elicits potassium efflux in mitochondria. Rupture of lysosomal membrane leads to cathepsin leakage into the cytosol.60, 61, 62 ROS, potassium efflux, cathepsins, and DAMPs could all be sensed by inflammasome sensors and cause inflammasome activation. Physiologically, the process of autophagy serves as a natural extinguisher for inflammasome. With the autophagic adaptor of p62, autophagosome recruits inflammasome components and promotes their degradation in a lysosome‐dependent manner. In addition, autophagy removes damaged mitochondria through autophagosomes, preventing the release of ROS and mitochondrial DNA into the cytoplasm.63, 64, 65 In the lesion of ischemic stroke, the autophagic process is suppressed, which further facilitates inflammasome activation65 (Figures 2, 3 and 4). Activation of inflammasome in microglia/macrophage exerts detrimental impacts on brain tissue undergoing ischemic injury.66 As that in chronic neurogenerative diseases, inflammasome serves as an amplifier of post‐stroke neural inflammation. A cascade of inflammatory reactions is induced by inflammasome products released by microglia/macrophage, contributing to the BBB breakdown and the subsequent leukocyte infiltration.67

Figure 2.

Figure 2

Inflammasome activation inhibits autophagy during aging. Autophagy can negatively regulate inflammasome activation by removing mitochondria‐derived stimuli. Aging impaired fusion of lysosomes with autophagosome, resulting in inflammasome activation by mitochondrial DNA (mtDNA) and ROS efflux from impaired mitochondria

Figure 3.

Figure 3

Vicious circle form between Aβ deposition and NLRP3 inflammasome activation in microglia/macrophage during AD. Inflammasome is activated in microglia/macrophage by Aβ directly, or by lysosomal damage and ROS production resulted from Aβ deposition. On the other hand, activation of inflammasome promotes Aβ aggregation exacerbating AD pathology

Figure 4.

Figure 4

Interactions of inflammasome in microphage with neural inflammation and aging‐related diseases. Inflammasome activation in microglia/macrophage could be triggered by Aβ plague deposition and tau neurofibrillary tangle in AD, α‐synuclein in PD, and neurons necroptosis in CNS stroke. Therefore, inflammasome activation is one of the consequences of CNS disease development. On the other hand, inflammasome activation exacerbates to neural inflammation, which plays detrimental role in neurodegenerative diseases and CNS stroke. Thus, inflammasome activation is one of the reasons for CNS disease progression

Accumulative evidences indicate that targeting inflammasome pharmacologically may salvage penumbral tissue in cerebral ischemia.68, 69 Remarkably, the NLRP3 inhibitor MCC950 and the NLRP1 neutralizing antibodies have been proved to be curative in ischemic stroke.70 Besides, repressing inflammasome through enhancing autophagy has also been proved to offer protection in ischemic stroke.71 Inhibiting inflammasome to limit excessive neural inflammation in acute ischemic stroke should be a promising therapeutic strategy, which needs further efforts for clinical translation.

5.2. Inflammasome and hemorrhagic transformation after ischemic stroke

Hemorrhagic transformation (HT) is one of the major complications of ischemic stroke. Activation of inflammasome during ischemic stroke promotes the occurrence of hemorrhagic transformation. Inflammasome products initial a robust inflammatory cascade, which exacerbates BBB injury, thus leading to vessel rupture and intracerebral bleeding.72 Currently, thrombolytic therapy is the only permissive pharmacological intervention for ischemic stroke. Nevertheless, patients that receive thrombolytic therapy have to confront increased risk of HT. Therefore, a strict time window is set up (4.5 hours) for thrombolytic treatment.73 Recently, it has been found that delayed recombinant tissue plasminogen activator (rtPA) treatment after ischemic stroke increased inflammasome activation in microglia thus increasing the risk of HT after ischemic stroke, which sheds light on the conclusion that inflammasome activation is the key limitation for therapeutic window for rtPA.74 Inhibiting inflammasome activation has displayed preventive efficacy for HT in ischemic stroke. NLRP3 inhibitor, MCC950, reduced the rate of HT in stroke animal models, accompanied by decreased leukocyte recruitment.70, 75 The evidence indicates that inflammasome inhibiting treatment may help to liberalize the time window for thrombolytic therapy through preventing HT occurrence.

5.3. Inflammasome activation in acute hemorrhagic stroke

Primary intracerebral hemorrhage (ICH) is another important subtype of acute stroke. Inflammasome plays a critical role in the pathophysiology of ICH‐induced brain injury. It has been found that inflammasome activates rapidly in microglia after the onset of ICH. As detected by Ma et al,76 inflammasome components, including NLRP3, cleaved Caspase‐1, and cleaved IL‐1β, were upregulated within 3 hours after ICH. As products of blood degradation, iron and heme initiate inflammasome activation in ROS‐dependent manner.77 Mitochondrial dysfunction is proved to be the major trigger for inflammasome activation in microglia during ICH pathology.76 Besides, miR‐223 has been demonstrated to directly regulate NLRP3 expression in ICH.78 As that in acute ischemic stroke, inflammasome products exacerbate BBB injury and promote leukocyte recruitment, thus enlarging perihematomal edema and exacerbating brain injury.76, 79 Targeting NLRP3 inflammasome activation may be a promising therapeutic strategy for ICH.80, 81 The P2X7 receptor is upstream of NLRP3 activation, and its inhibition has a pronounced neuroprotective effect in an ICH rat model.80 Blocking Caspase‐1 signaling or NLRP3 inflammasome has been demonstrated to effectively reduce the inflammatory responses and improved disease outcomes in ICH.82, 83

Phagocytosis of hematoma by microglia/macrophage is important to remove hematoma and ameliorate neuroinflammation after ICH. It is reported that phagocytosis, with abundant ROS production, could activate NLRP3 inflammasome in microglia/macrophage.84, 85 However, the impact of inflammasome activation on microglia/macrophage phagocytic activities remains elusive. Inflammasome activation in microglia/macrophage represents a certain kind of proinflammatory activity. It is well recorded that the proinflammatory phenotype of microglia/macrophage displays downregulated phagocytic capacity. On the other hand, it is the inflammation‐resolving microglia/macrophage that bears the full capacity of phagocytosis. Therefore, we infer that inhibiting inflammasome activation could protect microglia/macrophage from the self‐injury during phagocytosis and keep microglia/macrophage in the inflammatory‐resolving phenotype which facilitates hematoma clearance.

6. TARGETING INFLAMMASOME TO RETARD THE PACE OF AGING AND BRAKE AGING‐RELATED CNS DISEASES

Aging and the associated disorders prime CNS‐resident microglia and infiltrated macrophage for inflammasome activation during “healthy” and disease status. Conversely, inflammasome accelerates the aging process and exacerbates aging‐associated CNS diseases. Metabolism disorder is frequently observed in the elderly. As mentioned, accumulation of multiple metabolites could trigger inflammasome activation.25, 27 The other way round, activation of inflammasome exacerbates the status of metabolic disorder. Inflammasome contributes to glucose homeostasis, drives catecholamine degradation, blunts lipolysis, which causes visceral adiposity, and decreases substrate mobilization in the aged individuals.86 Therefore, developing lifestyle could prevent inflammasome activation, thus retards the pace of aging. It is evident that increased caffeine intake is relevant to lower inflammasome activation.25 Caloric restriction or a low‐carbohydrate ketogenic diet could inhibit inflammasome activation through elevating the level of β‐hydroxybutyrate.87 Proper physical exercise would prevent the induction of inflammasome.88 Sound sleep also helps to keep inflammasome quiescent.89 Confronting the aging‐related CNS diseases, targeting inflammasome is a promising therapeutic strategy. A broad body of evidences suggest that inhibiting inflammasome activation displays favorable curative effects in neurodegenerative diseases 42, 50, 52 as well as acute CNS injuries.70, 75, 82, 83

7. CONCLUSION

Senescence and the concomitant “inflammaging” are both the reason and the result of CNS inflammasome activation. CNS‐resident microglia and peripheral macrophage are the main cell types in which inflammasome activation is initiated. In “healthy” aged brains and CNS with aging‐related diseases, inflammasome activation in microglia/macrophage is broadly observed. Preventing inflammasome activation by improving lifestyle should be favorable for delaying senility. Inhibiting inflammasome activation with pharmacological interventions may help put on the brake for aging‐related CNS diseases.

CONFLICT OF INTEREST

The authors declare no conflicts of interest.

AUTHOR CONTRIBUTION

MH and YL wrote the manuscript. BZ and DL conducted the manuscript editing. WC and ZL designed and critically revised the manuscript. All authors read and approved the final manuscript.

ACKNOWLEDGMENTS

Not applicable.

Hu M‐Y, Lin Y‐Y, Zhang B‐J, Lu D‐L, Lu Z‐Q, Cai W. Update of inflammasome activation in microglia/macrophage in aging and aging‐related disease. CNS Neurosci Ther. 2019;25:1299–1307. 10.1111/cns.13262

M. Hu and Y. Lin are contributed equally to this work.

Funding information

This work was supported by the China Postdoctoral Science Foundation Grant (2018M643332) and the Chinese Natural Science Foundation (NCSF) grants (81671178 to Z. L).

Contributor Information

Zheng‐qi Lu, Email: lzq1828@outlook.com.

Wei Cai, Email: jchoigz@outlook.com.

REFERENCES

  • 1. Gabuzda D, Yankner BA. Physiology: inflammation links ageing to the brain. Nature. 2013;497(7448):197‐198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Sochocka M, Diniz BS, Leszek J. Inflammatory Response in the CNS: friend or Foe? Mol Neurobiol. 2017;54(10):8071‐8089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677‐687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Knezevic D, Verhoeff NPLG, Hafizi S, et al. Imaging microglial activation and amyloid burden in amnestic mild cognitive impairment. J Cereb Blood Flow Metab. 2018;38(11):1885‐1895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Fowler JH, McQueen J, Holland PR, et al. Dimethyl fumarate improves white matter function following severe hypoperfusion: involvement of microglia/macrophages and inflammatory mediators. J Cereb Blood Flow Metab. 2018;38(8):1354‐1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Takizawa T, Qin T, Lopes de Morais A, et al. Non‐invasively triggered spreading depolarizations induce a rapid pro‐inflammatory response in cerebral cortex. J Cereb Blood Flow Metab. 2019:271678X19859381 10.1177/0271678X19859381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Robblee MM, Kim CC, Porter Abate J, et al. Saturated fatty acids engage an IRE1alpha‐dependent pathway to activate the NLRP3 inflammasome in myeloid cells. Cell Rep. 2016;14(11):2611‐2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Zhu J, Hu Z, Han X, et al. Dopamine D2 receptor restricts astrocytic NLRP3 inflammasome activation via enhancing the interaction of beta‐arrestin2 and NLRP3. Cell Death Differ. 2018;25(11):2037‐2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. McKenzie BA, Mamik MK, Saito LB, et al. Caspase‐1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc Natl Acad Sci USA. 2018;115(26):E6065‐E6074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Adamczak SE, de Rivero Vaccari JP, Dale G, et al. Pyroptotic neuronal cell death mediated by the AIM2 inflammasome. J Cereb Blood Flow Metab. 2014;34(4):621‐629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Tsai YM, Chiang KH, Hung JY, et al. Der f1 induces pyroptosis in human bronchial epithelia via the NLRP3 inflammasome. Int J Mol Med. 2018;41(2):757‐764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. L'homme L, Esser N, Riva L, et al. Unsaturated fatty acids prevent activation of NLRP3 inflammasome in human monocytes/macrophages. J Lipid Res. 2013;54(11):2998‐3008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Minkiewicz J, de Rivero Vaccari JP, Keane RW. Human astrocytes express a novel NLRP2 inflammasome. Glia. 2013;61(7):1113‐1121. [DOI] [PubMed] [Google Scholar]
  • 14. He Y, Hara H, Nunez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012‐1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. He Y, Zeng MY, Yang D, Motro B, Nunez G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature. 2016;530(7590):354‐357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Xu J, Lu L, Li L. NEK7: a novel promising therapy target for NLRP3‐related inflammatory diseases. Acta Biochim Biophys Sin (Shanghai). 2016;48(10):966‐968. [DOI] [PubMed] [Google Scholar]
  • 17. Halle A, Hornung V, Petzold GC, et al. The NALP3 inflammasome is involved in the innate immune response to amyloid‐beta. Nat Immunol. 2008;9(8):857‐865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008;320(5876):674‐677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Shi J, Zhao Y, Wang Y, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014;514(7521):187‐192. [DOI] [PubMed] [Google Scholar]
  • 20. Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660‐665. [DOI] [PubMed] [Google Scholar]
  • 21. Hoare M, Narita M. Transmitting senescence to the cell neighbourhood. Nat Cell Biol. 2013;15(8):887‐889. [DOI] [PubMed] [Google Scholar]
  • 22. Yan T, Chen Z, Chopp M, et al. Inflammatory responses mediate brain‐heart interaction after ischemic stroke in adult mice. J Cereb Blood Flow Metab. 2018:271678X18813317 10.1177/0271678X18813317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Linville RM, DeStefano JG, Sklar MB, Chu C, Walczak P, Searson PC. Modeling hyperosmotic blood‐brain barrier opening within human tissue‐engineered in vitro brain microvessels. J Cereb Blood Flow Metab. 2019:271678X19867980 10.1177/0271678X19867980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Calder PC, Bosco N, Bourdet‐Sicard R, et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res Rev. 2017;40:95‐119. [DOI] [PubMed] [Google Scholar]
  • 25. Furman D, Chang J, Lartigue L, et al. Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states. Nat Med. 2017;23(2):174‐184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Mejias NH, Martinez CC, Stephens ME, de Rivero Vaccari JP. Contribution of the inflammasome to inflammaging. J Inflamm (Lond). 2018;15:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Youm Y‐H, Grant R, McCabe L, et al. Canonical Nlrp3 inflammasome links systemic low‐grade inflammation to functional decline in aging. Cell Metab. 2013;18(4):519‐532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Rippo MR, Olivieri F, Monsurro V, Prattichizzo F, Albertini MC, Procopio AD. MitomiRs in human inflamm‐aging: a hypothesis involving miR‐181a, miR‐34a and miR‐146a. Exp Gerontol. 2014;56:154‐163. [DOI] [PubMed] [Google Scholar]
  • 29. Chang CC, Li HH, Chang YT, et al. Abeta exacerbates alpha‐synuclein‐induced neurotoxicity through impaired insulin signaling in alpha‐synuclein‐overexpressed human SK‐N‐MC neuronal cells. CNS Neurosci Ther. 2018;24(1):47‐57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Jabir NR, Khan FR, Tabrez S. Cholinesterase targeting by polyphenols: a therapeutic approach for the treatment of Alzheimer's disease. CNS Neurosci Ther. 2018;24(9):753‐762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Parajuli B, Sonobe Y, Horiuchi H, Takeuchi H, Mizuno T, Suzumura A. Oligomeric amyloid beta induces IL‐1beta processing via production of ROS: implication in Alzheimer's disease. Cell Death Dis. 2013;4:e975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Labzin LI, Heneka MT, Latz E. Innate immunity and neurodegeneration. Annu Rev Med. 2018;69:437‐449. [DOI] [PubMed] [Google Scholar]
  • 33. Salminen A, Ojala J, Suuronen T, Kaarniranta K, Kauppinen A. Amyloid‐beta oligomers set fire to inflammasomes and induce Alzheimer's pathology. J Cell Mol Med. 2008;12(6A):2255‐2262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Huang Y, Huang X, Zhang L, et al. Magnesium boosts the memory restorative effect of environmental enrichment in Alzheimer's disease mice. CNS Neurosci Ther. 2018;24(1):70‐79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Nakanishi A, Kaneko N, Takeda H, et al. Amyloid beta directly interacts with NLRP3 to initiate inflammasome activation: identification of an intrinsic NLRP3 ligand in a cell‐free system. Inflamm Regen. 2018;38:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Venegas C, Kumar S, Franklin BS, et al. Microglia‐derived ASC specks cross‐seed amyloid‐beta in Alzheimer's disease. Nature. 2017;552(7685):355‐361. [DOI] [PubMed] [Google Scholar]
  • 37. Déniz‐Naranjo MC, Muñoz‐Fernandez C, Alemany‐Rodríguez MJ, et al. Cytokine IL‐1 beta but not IL‐1 alpha promoter polymorphism is associated with Alzheimer disease in a population from the Canary Islands, Spain. Eur J Neurol. 2008;15(10):1080‐1084. [DOI] [PubMed] [Google Scholar]
  • 38. Rubio‐Perez JM, Morillas‐Ruiz JM. A review: inflammatory process in Alzheimer's disease, role of cytokines. Sci World J. 2012;2012:756357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Feng J, Wang JX, Du YH, et al. Dihydromyricetin inhibits microglial activation and neuroinflammation by suppressing NLRP3 inflammasome activation in APP/PS1 transgenic mice. CNS Neurosci Ther. 2018;24(12):1207‐1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zoufal V, Wanek T, Krohn M, et al. Age dependency of cerebral P‐glycoprotein function in wild‐type and APPPS1 mice measured with PET. J Cereb Blood Flow Metab. 2018:271678X18806640 10.1177/0271678X18806640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Heneka MT, Kummer MP, Stutz A, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674‐678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Mawhinney LJ, de Rivero Vaccari JP, Dale GA, Keane RW, Bramlett HM. Heightened inflammasome activation is linked to age‐related cognitive impairment in Fischer 344 rats. BMC Neurosci. 2011;12:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Wu JY, Zhang Y, Wu WB, Hu G, Xu Y. Impaired long contact white matter fibers integrity is related to depression in Parkinson's disease. CNS Neurosci Ther. 2018;24(2):108‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Chen HM, Sha ZQ, Ma HZ, He Y, Feng T. Effective network of deep brain stimulation of subthalamic nucleus with bimodal positron emission tomography/functional magnetic resonance imaging in Parkinson's disease. CNS Neurosci Ther. 2018;24(2):135‐143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Huang J‐Z, Ren YI, Xu Y, et al. The delta‐opioid receptor and Parkinson's disease. CNS Neurosci Ther. 2018;24(12):1089‐1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhou Y, Lu M, Du R‐H, et al. MicroRNA‐7 targets Nod‐like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson's disease. Mol Neurodegener. 2016;11:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Sarkar S, Malovic E, Harishchandra DS, et al. Mitochondrial impairment in microglia amplifies NLRP3 inflammasome proinflammatory signaling in cell culture and animal models of Parkinson's disease. NPJ Parkinsons Dis. 2017;3:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Li D, Yang H, Ma J, Luo S, Chen S, Gu Q. MicroRNA‐30e regulates neuroinflammation in MPTP model of Parkinson's disease by targeting Nlrp3. Hum Cell. 2018;31(2):106‐115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Yan Y, Jiang W, Liu L, et al. Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell. 2015;160(1–2):62‐73. [DOI] [PubMed] [Google Scholar]
  • 50. Gordon R, Albornoz EA, Christie DC, et al. Inflammasome inhibition prevents alpha‐synuclein pathology and dopaminergic neurodegeneration in mice. Sci Transl Med. 2018;10(465):pii: eaah4066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Codolo G, Plotegher N, Pozzobon T, et al. Triggering of inflammasome by aggregated alpha‐synuclein, an inflammatory response in synucleinopathies. PLoS ONE. 2013;8(1):e55375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Hung KC, Huang HJ, Wang YT, Lin AM. Baicalein attenuates alpha‐synuclein aggregation, inflammasome activation and autophagy in the MPP(+)‐treated nigrostriatal dopaminergic system in vivo. J Ethnopharmacol. 2016;194:522‐529. [DOI] [PubMed] [Google Scholar]
  • 53. Kelly‐Hayes M. Influence of age and health behaviors on stroke risk: lessons from longitudinal studies. J Am Geriatr Soc. 2010;58(Suppl 2):S325‐S328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Yousufuddin M, Young N. Aging and ischemic stroke. Aging (Albany NY). 2019;11(9):2542‐2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Orfila JE, Dietz RM, Rodgers KM, et al. Experimental pediatric stroke shows age‐specific recovery of cognition and role of hippocampal Nogo‐A receptor signaling. J Cereb Blood Flow Metab. 2019:271678X19828581 10.1177/0271678X19828581 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Ritzel RM, Lai Y‐J, Crapser JD, et al. Aging alters the immunological response to ischemic stroke. Acta Neuropathol. 2018;136(1):89‐110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Boehme AK, Esenwa C, Elkind MS. Stroke risk factors, genetics, and prevention. Circ Res. 2017;120(3):472‐495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Iyer SS, Pulskens WP, Sadler JJ, et al. Necrotic cells trigger a sterile inflammatory response through the Nlrp3 inflammasome. Proc Natl Acad Sci USA. 2009;106(48):20388‐20393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Savage CD, Lopez‐Castejon G, Denes A, Brough D. NLRP3‐inflammasome activating DAMPs stimulate an inflammatory response in Glia in the absence of priming which contributes to brain inflammation after injury. Front Immunol. 2012;3:288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Hornung V, Bauernfeind F, Halle A, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9(8):847‐856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Bruchard M, Mignot G, Derangère V, et al. Chemotherapy‐triggered cathepsin B release in myeloid‐derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat Med. 2013;19(1):57‐64. [DOI] [PubMed] [Google Scholar]
  • 62. Alishahi M, Farzaneh M, Ghaedrahmati F, Nejabatdoust A, Sarkaki A, Khoshnam SE. NLRP3 inflammasome in ischemic stroke: as possible therapeutic target. Int J Stroke. 2019;14(6):574‐591. [DOI] [PubMed] [Google Scholar]
  • 63. Harris J, Hartman M, Roche C, et al. Autophagy controls IL‐1beta secretion by targeting pro‐IL‐1beta for degradation. J Biol Chem. 2011;286(11):9587‐9597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Shi CS, Shenderov K, Huang NN, et al. Activation of autophagy by inflammatory signals limits IL‐1beta production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255‐263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Gao L, Dong Q, Song Z, Shen F, Shi J, Li Y. NLRP3 inflammasome: a promising target in ischemic stroke. Inflamm Res. 2017;66(1):17‐24. [DOI] [PubMed] [Google Scholar]
  • 66. He X‐F, Zeng Y‐X, Li GE, et al. Extracellular ASC exacerbated the recurrent ischemic stroke in an NLRP3‐dependent manner. J Cereb Blood Flow Metab. 2019:271678X19856226 10.1177/0271678X19856226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Miners JS, Schulz I, Love S. Differing associations between Abeta accumulation, hypoperfusion, blood‐brain barrier dysfunction and loss of PDGFRB pericyte marker in the precuneus and parietal white matter in Alzheimer's disease. J Cereb Blood Flow Metab. 2018;38(1):103‐115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Hong PU, Gu R‐N, Li F‐X, et al. NLRP3 inflammasome as a potential treatment in ischemic stroke concomitant with diabetes. J Neuroinflammation. 2019;16(1):121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Zhang N, Zhang X, Liu X, et al. Chrysophanol inhibits NALP3 inflammasome activation and ameliorates cerebral ischemia/reperfusion in mice. Mediators Inflamm. 2014;2014:370530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Ismael S, Zhao L, Nasoohi S, Ishrat T. Inhibition of the NLRP3‐inflammasome as a potential approach for neuroprotection after stroke. Sci Rep. 2018;8(1):5971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. He Q, Li Z, Wang Y, Hou Y, Li L, Zhao J. Resveratrol alleviates cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasome activation through Sirt1‐dependent autophagy induction. Int Immunopharmacol. 2017;50:208‐215. [DOI] [PubMed] [Google Scholar]
  • 72. Cao G, Jiang N, Hu Y, et al. Ruscogenin attenuates cerebral ischemia‐induced blood‐brain barrier dysfunction by suppressing TXNIP/NLRP3 inflammasome activation and the MAPK pathway. Int J Mol Sci. 2016;17(9):pii:E1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Powers WJ, Rabinstein AA, Ackerson T, et al. 2018 Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2018;49(3):e46‐e110. [DOI] [PubMed] [Google Scholar]
  • 74. Guo Z, Yu S, Chen X, et al. Suppression of NLRP3 attenuates hemorrhagic transformation after delayed rtPA treatment in thromboembolic stroke rats: Involvement of neutrophil recruitment. Brain Res Bull. 2018;137:229‐240. [DOI] [PubMed] [Google Scholar]
  • 75. Lemarchand E, Barrington J, Chenery A, et al. Extent of ischemic brain injury after thrombotic stroke is independent of the NLRP3 (NACHT, LRR and PYD domains‐containing protein 3) inflammasome. Stroke. 2019;50(5):1232‐1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ma Q, Chen S, Hu Q, Feng H, Zhang JH, Tang J. NLRP3 inflammasome contributes to inflammation after intracerebral hemorrhage. Ann Neurol. 2014;75(2):209‐219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Qu J, Chen W, Hu R, Feng H. The injury and therapy of reactive oxygen species in intracerebral hemorrhage looking at mitochondria. Oxid Med Cell Longev. 2016;2016:2592935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Yang Z, Zhong L, Xian R, Yuan B. MicroRNA‐223 regulates inflammation and brain injury via feedback to NLRP3 inflammasome after intracerebral hemorrhage. Mol Immunol. 2015;65(2):267‐276. [DOI] [PubMed] [Google Scholar]
  • 79. Aronowski J, Hall CE. New horizons for primary intracerebral hemorrhage treatment: experience from preclinical studies. Neurol Res. 2005;27(3):268‐279. [DOI] [PubMed] [Google Scholar]
  • 80. Feng L, Chen Y, Ding R, et al. P2X7R blockade prevents NLRP3 inflammasome activation and brain injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite. J Neuroinflammation. 2015;12:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Shao BZ, Cao Q, Liu C. Targeting NLRP3 inflammasome in the treatment of CNS diseases. Front Mol Neurosci. 2018;11:320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Wu B, Ma Q, Khatibi N, et al. Ac‐YVAD‐CMK decreases blood‐brain barrier degradation by inhibiting caspase‐1 activation of interleukin‐1beta in intracerebral hemorrhage mouse model. Transl Stroke Res. 2010;1(1):57‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Ren H, Kong Y, Liu Z, et al. Selective NLRP3 (pyrin domain‐containing protein 3) inflammasome inhibitor reduces brain injury after intracerebral hemorrhage. Stroke. 2018;49(1):184‐192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Yang Y, Wang H, Kouadir M, Song H, Shi F. Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis. 2019;10(2):128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Meissner F, Seger RA, Moshous D, Fischer A, Reichenbach J, Zychlinsky A. Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease. Blood. 2010;116(9):1570‐1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Mandrup‐Poulsen T. Immunometabolism in 2017: metabolism and the inflammasome in health and ageing. Nat Rev Endocrinol. 2018;14(2):72‐74. [DOI] [PubMed] [Google Scholar]
  • 87. Shao BZ, Xu ZQ, Han BZ, Su DF, Liu C. NLRP3 inflammasome and its inhibitors: a review. Front Pharmacol. 2015;6:262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Wang Y, Xu Y, Sheng H, Ni X, Lu J. Exercise amelioration of depression‐like behavior in OVX mice is associated with suppression of NLRP3 inflammasome activation in hippocampus. Behav Brain Res. 2016;307:18‐24. [DOI] [PubMed] [Google Scholar]
  • 89. Li X, Liang S, Li Z, et al. Leptin increases expression of 5‐HT2B receptors in astrocytes thus enhancing action of fluoxetine on the depressive behavior induced by sleep deprivation. Front Psychiatry. 2018;9:734. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES