REVIEW ARTICLE


Inflammasome can Affect Adult Neurogenesis: A Review Article



Sara Solimani Asl1, Cyrus Jalili2, Tayebeh Artimani1, Mahdi Ramezani1, Fatemeh Mirzaei3, *
iD

1 Endometrium and Endometriosis Research Centre, Hamadan University of Medical Sciences, Hamadan, Iran
2 Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
3 Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran


Article Metrics

CrossRef Citations:
0
Total Statistics:

Full-Text HTML Views: 1405
Abstract HTML Views: 643
PDF Downloads: 405
ePub Downloads: 256
Total Views/Downloads: 2709
Unique Statistics:

Full-Text HTML Views: 660
Abstract HTML Views: 287
PDF Downloads: 296
ePub Downloads: 170
Total Views/Downloads: 1413



Creative Commons License
© 2021 Asl et al.

open-access license: This is an open access article distributed under the terms of the Creative Commons Attribution 4.0 International Public License (CC-BY 4.0), a copy of which is available at: https://creativecommons.org/licenses/by/4.0/legalcode. This license permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

* Address correspondence to this author at the Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; E-mail: fmirzaei90@yahoo.com


Abstract

Adult neurogenesis is the process of producing new neurons in the adult brain and is limited to two major areas: the hippocampal dentate gyrus and the Subventricular Zone (SVZ). Adult neurogenesis is affected by some physiological, pharmacological, and pathological factors. The inflammasome is a major signalling platform that regulates caspase-1 and induces proinflammatory cytokines production such as interleukin-1β (IL1-β) and IL-18.

Inflammasomes may be stimulated through multiple signals, and some of these signaling factors can affect neurogenesis. In the current review, “adult neurogenesis and inflammasome” were searched in PubMed, Scopus, and Google Scholar. Reviewing various research works showed correlations between inflammasome and neurogenesis by different intermediate factors, such as interferons (IFN), interleukins (IL), α-synuclein, microRNAs, and natural compounds. Concerning the significant role of neurogenesis in the health of the nervous system and memory, understanding factors inducing neurogenesis is crucial for identifying new therapeutic aims. Hence in this review, we will discuss the different mechanisms by which inflammasome influences adult neurogenesis.

Keywords: Inflammasome, Neurogenesis, Neural stem/progenitor cells, Subgranular zone, Subventricular zone, Hippocampal dentate gyrus.



1. INTRODUCTION AND STATEMENT OF THE PROBLEM

Throughout life, neural stem/progenitor cells [NSPCs] can produce new neurons in specific areas in the mammalian’s brain [1]. Adult neurogenesis contributes to physiological brain activities and is crucial for specific learning and memory processes and a critical factor in adult brain plasticity. Adult neurogenesis is limited to the Subventricular Zone (SVZ) and the hippocampal Dentate Gyrus [2] (Fig. 1).

Adult neurogenesis is an active process done with high accuracy and can be affected by pharmacological intervention, different physiological and pathological conditions [3]. Inflammasomes are the main signalling platforms that recognize sterile factors and pathogenic microorganisms [4]. During inflammasome activation, caspase-1, the prominent inflammatory mediator [5], releases interleukin-18 and interleukin-1β (IL-18/ IL1-β), and these products cause pyroptosis and cells death [6, 7]. The innate immune system

cells express Pattern Recognition Receptors (PRRs) to specify two molecular classes: Pathogen-Associated Molecular Patterns (PAMPs) and Damage-Associated Molecular Patterns (DAMPs). NOD-Like Receptors (NLRs) are a subclass of PRRs which are found in the cytosol. There are numerous NLRs proteins that contribute to inflammasome pathways [8]. The oligomerization of NLRs (NLRP1, NLRP3, and NLRC4) can form multi-protein inflammasome complexes [9] (Table 1).

Toll-like Receptors (TLRs) are PRRs on immune cells which detect pathogens and stimulate IL1-β production [10] (Fig. 2). As inflammasome causes pyroptosis, it is possible that inflammasome may reduce neurogenesis. Several studies showed that IL1-β, the product of inflammation and IL-6, and the downstream target of IL1-β, can potentially prevent neurogenesis [2]. IL1-β, a potent pro-inflammatory cytokine, is important for host-defense reactions to injury and infection and is released and produced by many cell types [11]. It was reported that IL1-β had preventive effects on hippocampal cells in the dentate gyrus and NPCs of adult rats [12]. According to the Ryan study, IL1-β reduced neurogenesis and proliferation, while co-treatment with IL1-β receptor antagonist inhibited the adverse impact of IL1-β on the proliferation of cells [13].

Fig. (1). Zones of adult neurogenesis: SVZ; Sub Ventricular Zone/ SGZ; Sub Granular Zone/ NSC; Neural Stem Cell.

Fig. (2). Inflammasome: PAMPs; pathogen-associated molecular patterns/ DAMPs; damage-associated molecular patterns / TLRs; Toll like receptors /NLRS; NOD-Like Receptors /IL; Interleukin / NF-κB; nuclear factor kappa B.

Table 1. Different factors responsible for active or inactive inflammasome and affect neurogenesis.
Effect on Neurogenesis Role in Inflammasome Factor
Inhibits neurogenesis It is produced by activation of the inflammasome IL1β
Inhibits neurogenesis It is a downstream target of IL1β IL6
Stimulates neurogenesis Inhibits IL1β, IL6, and TNF-α IL4
Stimulates neurogenesis Inhibits NLRP3 inflammasome IL10
Inhibits neurogenesis It is produced by activation of the inflammasome IL18
Inhibits neurogenesis Activates inflammasome INF-α
Its role is dose-dependent In the low dose, it inhibits the activation of NLRP1 inflammasome.
Activates inflammasome at high doses
INF- β
Stimulates neurogenesis Inhibits α-synuclein and inhibits NLRP3 inflammasome miR-7
Stimulates neurogenesis Inhibits NLRP3 inflammasome miR-9
Stimulates neurogenesis Inhibits NLRP3 inflammasome miR-223
Stimulates neurogenesis Inhibits TXNIP that is one activator of NLRP3 inflammasome Curcumin
Stimulates neurogenesis Inhibits NLRP3 inflammasome EGCG
Stimulates neurogenesis Inhibits NLRP3 inflammasome Quercetin
Stimulates neurogenesis Inhibits NLRP3 inflammasome Boswellia serrate

IL1-β also decreased proliferation and differentiation of rat neonatal DG NPCs to serotonergic (5-hydroxytryptamine) neurons [14]. Since IL-1 β is the inflammasome's ultimate production, therefore neurogenesis may be enhanced by managing the inflammasome and reducing the IL1-β production.

Interleukin-6 (IL-6) is an IL1-β downstream target, increased continuously in patient’s serum with NLRP3 inflammasome mediated conditions [2]. IL-6 reduced neurogenesis and enhanced apoptosis in rat adult DG NPCs, blocking antibodies to IL-6 induced neurogenesis [15].

The crucial role of IL-18 is highlighted in mediating neurodegeneration and neuroinflammation in the CNS under pathological conditions [16]. IL-18 exhibits direct pro-inflammatory properties by increasing inflammatory factors such as TNF-α, IL1-β, and IL-6 [17]. As previously described, TNF-α, IL1-β, and IL-6 have inhibitory effects on neurogenesis. Also, IL-18 increases amyloid precursor protein [18], Beta-secretase 1 (BACE1), and the N-terminal fragment of presenilin-1 and presenilin enhancer-2 protein levels [19]. It has been reported that in the adult SGZ, presenilin (PS) variant expression is correlated to early-onset familial Alzheimer's [20]. So, IL-18 secreted during inflammasome influences neurogenesis not only by above mentioned inflammatory factors, but also via presenilin expression in adult SGZ.

Another interleukin that, unlike the expressed interleukins, has a protective effect against inflammation is IL-4. It inhibits inflammasome and reduces IL-6, IL1-β, and TNF-α [21]. In many studies, the effect of IL-4 on immunity has been shown. It is also prominent in the normal brain’s functions, involving learning, memory, and neurogenesis [22]. IL-4 increases neuronal and glial differentiation [14]. Changes in microglial status by IL-4 alter their phenotype to enhance neurogenesis [23, 24]. It can be said that IL-4 enhances neurogenesis via inhibiting inflammasome, IL1-β, TNF-α, and IL-6 and therefore increases neurogenesis. IL-10 also inhibits NLRP3 inflammasome and increases neurogenesis through the inhibition of IL1-β production [25]. It elevates proliferation and decreases neuronal and glial differentiation [14]. According to the Gurung et al. study, IL-10 controls NLRP3 inflammasome activation negatively [26]. It inhibits the signaling of nuclear factor kappa B [NF-κB] during inflammasome [27]. It also prevents TNF-α generation of human monocytes [28]. In neurons, this interleukin receptor signaling is correlated with elevated cell survival [29] and the regulation of adult neurogenesis [30].

Another effective factor in the inflammasome pathway is TLR. TLRs, as the main regulators of the innate immune system, are receptors that contributed to the inflammasome. The immune signal is activated through NF-κB to prime the NLRP3 inflammasome [31].

IFN-α and IFN-β are inflammasome products and control inflammasome activation [32].

Interferons α and β, in addition to being key players at the start of the inflammasome, are two of its ultimate products [33]. It has been assumed that IFN-α suppresses NSC proliferation directly and, as a result, reduces new neurons generation [34] and inhibits the proliferation of cells in adult rat’s SGZ [35]. Chronic peripheral IFN-α administration suppresses neurogenesis and induces depressive behavioral phenotypes [34]. The impact of IFN-β on neurogenesis depends on its concentration. Low IFN-β concentrations (1000 U/ml) contribute to the survival and proliferation of human NSC, while greater concentrations (more than 100,000 U/ml) reduce the survival and proliferation of human NSC [36]. Controlling IFN-α and IFN-β from various pathways, like inflammasome, can enhance neurogenesis. There are other factors that cause neurogenesis suppression by activating the inflammation. α-synuclein (α-syn) is a ubiquitous protein particularly observed in high amounts in the brain and is assumed to have the main role in the pathogenesis of Alzheimer’s and Parkinson’s disease and other neurodegenerative disorders [37]. In patients with dementia, levels of hippocampal α-syn have been reported to increase [38]. α-syn motivates IL1-β production in a process that is dependent on the NLRP3 inflammasome in monocytes [39]. A53T mutant α-syn inhibited proliferation and differentiation of adult NSC in SVZ through activation of NLRP3/caspase-1/IL1-β signalling axis. Extracellular α-syn may stimulate pathways of pro-inflammatory TLR4 in astrocytes [40]. TLR4 generates IL1-β by the activation of the inflammasome. IL1-β inhibits neurogenesis. A negative effect of α-syn has been detected on newly generated neurons, especially on their dendrite development and spine improvement [38]. Accumulation of α-syn may be involved in the neurodegenerative process via weakening neurogenesis and the existence of differentiating neural progeny [41]. It can be summarized that α-syn causes an inhibitory effect on neurogenesis via activation of the inflammasome. Studies have shown that some microRNA inhibits TLR3 and NLRP3 by hindering α-synuclein. MicroRNA7, microRNA223, and microRNA9 inhibit the activation of NLRP3 inflammasome in NSCs, resulting in increased neurogenesis [42-45]. miR-7 regulates NLRP3 and TLR4 expression in adult NSCs directly [46]. miR-7 inhibits the activation of the microglial NLRP3 inflammasome, while anti-miR-7 aggravates the activation of inflammasome in vitro [42]. miR-7 simulator’s injection into lateral ventricles prevented the activation of NLRP3 inflammasome and enhanced adult neurogenesis in mouse SVZ [46]. Furthermore, intracerebroventricular injection of miR-7 improved the SVZ neurogenesis impairment and promoted adult SVZ neurogenesis by reducing NLRP3 inflammasome [46].

It was shown that miR-223 reduced the NLRP3 activity [43, 44] and cell-autonomous suppression of miR-223 in the adult mice dentate gyrus NS/PCs caused a potential rise in immature neurons soma size, dendritic tree size, branch number per neuron, and complexity; however, neuronal migration in the dentate gyrus was not affected [45]. miR-9 can also prevent the NLRP3 inflammasome activation [47] and control NS/PC proliferation [45]. Levels of miR-9 expression increase across the transition from neuronal precursors to neurons during the differentiation of embryonic stem cells [48]. miR-7, miR-9, and miR-223 may increase neurogenesis by suppression of inflammasome.

Some natural compounds have stimulant impacts on neurogenesis [49-51]. Curcumin [52], Epigallocatechin-3-gallate (EGCG), Boswellia serrate [53], and quercetin are examples of these natural compounds that enhance neurogenesis. Curcumin can decrease the release of IL1-β from microglia, inhibit microglial activation, decrease stroke injury, activate neurogenesis in the hippocampus, and trigger neuronal protective mechanisms like heat shock protein [HSP] elevation [54]. Studies reported that HSP90 inhibition influences the stability of free NLRP3 and pro-IL1-β proteins [55]. Epigallocatechin-3-gallate [EGCG] is the major antioxidant in green tea. EGCG, by activating SHH and suppressing NLRP3 and NF-κB, inhibits IL1-β, TNF-α, and caspase1 and stimulates neurogenesis [56]. Tozser et al. showed that EGCG promotes hippocampal adult neurogenesis [57]. Treatment of EGCG markedly increased BrdU-labeled cells in hippocampal cultures, neural progenitor cell (NPC), and in the adult mice’s dentate gyrus. Other findings showed that EGCG prompts neurogenesis via reduction of IL1-β and SHH pathway and, SHH ameliorates neurogenesis by the suppression of the inflammation pathway.

Boswellia serrate resin extract, which has 3-O-acetyl-11-keto-b-boswellic acid (AKBA), possesses anti-inflammatory features and has significant therapeutic properties to cure inflammatory illnesses. It decreases IL1-β and TNF-α in a dose-dependent manner [58]. It also controls inflammasome, decreases inflammatory cytokines, and suppresses NF-κB p65, TLR-3, and TLR-4 protein expression [59]. Boswellic acid inhibits TNF-α and IL1-β release in monocytes [60] and inhibits IL-1, IL-6, TNFα, and NF-κB [61]. Hippocampal neurogenesis after administration of Boswellia extracts significantly increased [53]. Boswellia Serrata up-regulates Brain-Derived Neurotrophic Factor [BDNF]. BDNF regulates adult SVZ neurogenesis [61]. The role played by BDNF in controlling SVZ neurogenesis was primarily observed by Goldman et al. in the 1990s. They reported that rat SVZ-derived neuroblasts treated with BDNF in vitro survived for a long time [62].

CONCLUSION

It is concluded that activation of the inflammasome in the brain can inhibit adult neurogenesis, and drugs or compounds that inhibit the inflammasome platform can improve the neurogenesis process in adults by different pathways.

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

None.

CONFLICTS OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

Declared none.

REFERENCES

[1] Braun SM, Jessberger S. Adult neurogenesis and its role in neuropsychiatric disease, brain repair and normal brain function. Neuropathol Appl Neurobiol 2014; 40(1): 3-12.
[2] McGeough MD, Pena CA, Mueller JL, et al. Cutting edge: IL-6 is a marker of inflammation with no direct role in inflammasome-mediated mouse models. J Immunol 2012; 189(6): 2707-11.
[3] Ming GL, Song H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 2011; 70(4): 687-702.
[4] Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol 2013; 13(6): 397-411.
[5] Guo H, Callaway JB, Ting JP-Y. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med 2015; 21(7): 677-87.
[6] Okun E, Griffioen KJ, Mattson MP. Toll-like receptor signaling in neural plasticity and disease. Trends Neurosci 2011; 34(5): 269-81.
[7] Abbasi-Oshaghi E, Mirzaei F, Pourjafar M. NLRP3 inflammasome, oxidative stress, and apoptosis induced in the intestine and liver of rats treated with titanium dioxide nanoparticles: in vivo and in vitro study. Int J Nanomedicine 2019; 14: 1919-36.
[8] Zambetti LP, Mortellaro A. NLRPs, microbiota, and gut homeostasis: unravelling the connection. J Pathol 2014; 233(4): 321-30.
[9] Fukata M, Vamadevan AS, Abreu MT, Eds. Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in inflammatory disorders. Seminars in immunology Elsevier. 2009; 21(4): 242-53.
[10] Grishman EK, White PC, Savani RC. Toll-like receptors, the NLRP3 inflammasome, and interleukin-1β in the development and progression of type 1 diabetes. Pediatr Res 2012; 71(6): 626-32.
[11] Dinarello CA. Biologic basis for interleukin-1 in disease. Blood 1996; 87(6): 2095-147.
[12] Crampton SJ, Collins LM, Toulouse A, Nolan YM, O’Keeffe GW. Exposure of foetal neural progenitor cells to IL-1β impairs their proliferation and alters their differentiation - a role for maternal inflammation? J Neurochem 2012; 120(6): 964-73.
[13] Ryan SM, O’Keeffe GW, O’Connor C, Keeshan K, Nolan YM. Negative regulation of TLX by IL-1β correlates with an inhibition of adult hippocampal neural precursor cell proliferation. Brain Behav Immun 2013; 33: 7-13.
[14] Borsini A, Zunszain PA, Thuret S, Pariante CM. The role of inflammatory cytokines as key modulators of neurogenesis. Trends Neurosci 2015; 38(3): 145-57.
[15] Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science 2003; 302(5651): 1760-5.
[16] Felderhoff-Mueser U, Schmidt OI, Oberholzer A, Bührer C, Stahel PF. IL-18: a key player in neuroinflammation and neurodegeneration? Trends Neurosci 2005; 28(9): 487-93.
[17] Netea MG, Kullberg BJ, Verschueren I, Van Der Meer JW. Interleukin-18 induces production of proinflammatory cytokines in mice: no intermediate role for the cytokines of the tumor necrosis factor family and interleukin-1beta. Eur J Immunol 2000; 30(10): 3057-60.
[18] Choudhury NR, de Lima Alves F, de Andrés-Aguayo L, et al. Tissue-specific control of brain-enriched miR-7 biogenesis. Genes Dev 2013; 27(1): 24-38.
[19] Sutinen EM, Pirttilä T, Anderson G, Salminen A, Ojala JO. Pro-inflammatory interleukin-18 increases Alzheimer’s disease-associated amyloid-β production in human neuron-like cells. J Neuroinflammation 2012; 9(1): 199.
[20] Feng R, Rampon C, Tang YP, et al. Deficient neurogenesis in forebrain-specific presenilin-1 knockout mice is associated with reduced clearance of hippocampal memory traces. Neuron 2001; 32(5): 911-26.
[21] Hwang I, Yang J, Hong S, et al. Non-transcriptional regulation of NLRP3 inflammasome signaling by IL-4. Immunol Cell Biol 2015; 93(6): 591-9.
[22] Gadani SP, Cronk JC, Norris GT, Kipnis J. IL-4 in the brain: a cytokine to remember. J Immunol 2012; 189(9): 4213-9.
[23] Carpentier PA, Palmer TD. Immune influence on adult neural stem cell regulation and function. Neuron 2009; 64(1): 79-92.
[24] Butovsky O, Ziv Y, Schwartz A, et al. Microglia activated by IL-4 or IFN-gamma differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells. Mol Cell Neurosci 2006; 31(1): 149-60.
[25] Strle K, Zhou JH, Shen WH, et al. Interleukin-10 in the brain. Crit Rev Immunol 2001; 21(5): 427-49.
[26] Gurung P, Li B, Subbarao Malireddi RK, Lamkanfi M, Geiger TL, Kanneganti TD. Chronic TLR stimulation controls NLRP3 inflammasome activation through IL-10 mediated regulation of NLRP3 expression and caspase-8 activation. Sci Rep 2015; 5: 14488.
[27] Greenhill CJ, Jones GW, Nowell MA, et al. Interleukin-10 regulates the inflammasome-driven augmentation of inflammatory arthritis and joint destruction. Arthritis Res Ther 2014; 16(4): 419.
[28] Dagvadorj J, Naiki Y, Tumurkhuu G, et al. Interleukin-10 inhibits tumor necrosis factor-alpha production in lipopolysaccharide-stimulated RAW 264.7 cells through reduced MyD88 expression. Innate Immun 2008; 14(2): 109-15.
[29] Zhou Z, Peng X, Insolera R, Fink DJ, Mata M. Interleukin-10 provides direct trophic support to neurons. J Neurochem 2009; 110(5): 1617-27.
[30] McPherson CA, Aoyama M, Harry GJ. Interleukin (IL)-1 and IL-6 regulation of neural progenitor cell proliferation with hippocampal injury: differential regulatory pathways in the subgranular zone (SGZ) of the adolescent and mature mouse brain. Brain Behav Immun 2011; 25(5): 850-62.
[31] Coutinho-Silva R, Ojcius DM. Role of extracellular nucleotides in the immune response against intracellular bacteria and protozoan parasites. Microbes Infect 2012; 14(14): 1271-7.
[32] Masters SL, Gerlic M, Metcalf D, et al. NLRP1 inflammasome activation induces pyroptosis of hematopoietic progenitor cells. Immunity 2012; 37(6): 1009-23.
[33] Guarda G, Braun M, Staehli F, et al. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity 2011; 34(2): 213-23.
[34] Zheng LS, Hitoshi S, Kaneko N, et al. Mechanisms for interferon-α-induced depression and neural stem cell dysfunction. Stem Cell Reports 2014; 3(1): 73-84.
[35] Kaneko N, Kudo K, Mabuchi T, et al. Suppression of cell proliferation by interferon-alpha through interleukin-1 production in adult rat dentate gyrus. Neuropsychopharmacology 2006; 31(12): 2619-26.
[36] Sun X, Shi L, Zhang H, Li R, Liang R, Liu Z. Effects of Toll-like receptor 3 on herpes simplex virus type-1-infected mouse neural stem cells. Can J Microbiol 2015; 61(3): 201-8.
[37] Kim S, Seo JH, Suh YH. Alpha-synuclein, Parkinson’s disease, and Alzheimer’s disease. Parkinsonism Relat Disord 2004; 10(Suppl. 1): S9-S13.
[38] Winner B, Regensburger M, Schreglmann S, et al. Role of α-synuclein in adult neurogenesis and neuronal maturation in the dentate gyrus. J Neurosci 2012; 32(47): 16906-16.
[39] Du RH, Tan J, Yan N, et al. Kir6.2 knockout aggravates lipopolysaccharide-induced mouse liver injury via enhancing NLRP3 inflammasome activation. J Gastroenterol 2014; 49(4): 727-36.
[40] Rannikko EH, Weber SS, Kahle PJ. Exogenous α-synuclein induces toll-like receptor 4 dependent inflammatory responses in astrocytes. BMC Neurosci 2015; 16: 57.
[41] Crews L, Mizuno H, Desplats P, et al. Alpha-synuclein alters Notch-1 expression and neurogenesis in mouse embryonic stem cells and in the hippocampus of transgenic mice. J Neurosci 2008; 28(16): 4250-60.
[42] Zhou Y, Lu M, Du RH, 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.
[43] Qazi O, Parthasarathy PT, Lockey R, Kolliputi N. Can microRNAs keep inflammasomes in check? Front Genet 2013; 4: 30.
[44] Bauernfeind F, Rieger A, Schildberg FA, Knolle PA, Schmid-Burgk JL, Hornung V. NLRP3 inflammasome activity is negatively controlled by miR-223. J Immunol 2012; 189(8): 4175-81.
[45] Harraz MM, Xu JC, Guiberson N, Dawson TM, Dawson VL. MiR-223 regulates the differentiation of immature neurons. Mol Cell Ther 2014; 2(18): 18.
[46] Fan Z, Lu M, Qiao C, Zhou Y, Ding JH, Hu G. MicroRNA-7 enhances subventricular zone neurogenesis by inhibiting NLRP3/Caspase-1 axis in adult neural stem cells. Mol Neurobiol 2016; 53(10): 7057-69.
[47] Wang Y, Han Z, Fan Y, et al. MicroRNA-9 inhibits nlrp3 inflammasome activation in human atherosclerosis inflammation cell models through the JAK1/STAT signaling pathway. Cell Physiol Biochem 2017; 41(4): 1555-71.
[48] Wakabayashi T, Hidaka R, Fujimaki S, Asashima M, Kuwabara T. MicroRNAs and epigenetics in adult neurogenesis. Adv Genet 2014; 86: 27-44.
[49] Mirzaei F, Khazaei M, Komaki A, Amiri I, Jalili C. Virgin coconut oil (VCO) by normalizing NLRP3 inflammasome showed potential neuroprotective effects in Amyloid-β induced toxicity and high-fat diet fed rat. Food Chem Toxicol 2018; 118: 68-83.
[50] Zargooshnia S, Shahidi S, Ghahremanitamadon F, Nikkhah A, Mehdizadeh M, Soleimani Asl S. The protective effect of Borago Officinalis extract on amyloid β (25-35)-induced long term potentiation disruption in the dentate gyrus of male rats. Metab Brain Dis 2015; 30(1): 151-6.
[51] Mehdizadeh M, Hashem Dabaghian F, Shojaee A, et al. Protective effects of cyperus rotundus extract on amyloid β-peptide (1-40)-induced memory impairment in male rats: a behavioral study. Basic Clin Neurosci 2017; 8(3): 249-54.
[52] Jalili C, Rodsari BA, Roshankhah S, et al. Effect of curcumin on hippocampus dentate gyrus injury induced by nicotine in rats. Journal of Herbmed Pharmacology 2019; 8(4): 320-7.
[53] Jalili C, Salahshoor MR, Pourmotabbed A, et al. The effects of aqueous extract of Boswellia Serrata on hippocampal region CA1 and learning deficit in kindled rats. Res Pharm Sci 2014; 9(5): 351-8.
[54] Xu Y, Ku B, Tie L, et al. Curcumin reverses the effects of chronic stress on behavior, the HPA axis, BDNF expression and phosphorylation of CREB. Brain Res 2006; 1122(1): 56-64.
[55] Bradley E, Zhao X, Wang R, Brann D, Bieberich E, Wang G. Low dose Hsp90 inhibitor 17AAG protects neural progenitor cells from ischemia induced death. J Cell Commun Signal 2014; 8(4): 353-62.
[56] Wang Y, Li M, Xu X, Song M, Tao H, Bai Y. Green tea epigallocatechin-3-gallate (EGCG) promotes neural progenitor cell proliferation and sonic hedgehog pathway activation during adult hippocampal neurogenesis. Mol Nutr Food Res 2012; 56(8): 1292-303.
[57] Tőzsér J, Benkő S. Natural Compounds as Regulators of NLRP3 Inflammasome-Mediated IL-1β Production. Mediators Inflamm 2016; 2016: 5460302.
[58] Sengupta K, Kolla JN, Krishnaraju AV, et al. Cellular and molecular mechanisms of anti-inflammatory effect of Aflapin: a novel Boswellia serrata extract. Mol Cell Biochem 2011; 354(1-2): 189-97.
[59] Chen LC, Hu LH, Yin MC. Alleviative effects from boswellic acid on acetaminophen-induced hepatic injury - Corrected and republished from: Biomedicine (Taipei) 2017; 6(2): 9.
[60] Syrovets T, Büchele B, Krauss C, Laumonnier Y, Simmet T. Acetyl-boswellic acids inhibit lipopolysaccharide-mediated TNF-alpha induction in monocytes by direct interaction with IkappaB kinases. J Immunol 2005; 174(1): 498-506.
[61] Siddiqui MZ. Boswellia serrata, a potential antiinflammatory agent: an overview. Indian J Pharm Sci 2011; 73(3): 255-61.
[62] Kirschenbaum B, Goldman SA. Brain-derived neurotrophic factor promotes the survival of neurons arising from the adult rat forebrain subependymal zone. Proc Natl Acad Sci USA 1995; 92(1): 210-4.