Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1694 word(s) 1694 2021-06-29 10:04:01 |
2 format correction Meta information modification 1694 2021-07-07 03:15:04 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tarakcioglu, E. Melatonin on NLRP3 Inflammasome Activation. Encyclopedia. Available online: https://encyclopedia.pub/entry/11726 (accessed on 16 April 2024).
Tarakcioglu E. Melatonin on NLRP3 Inflammasome Activation. Encyclopedia. Available at: https://encyclopedia.pub/entry/11726. Accessed April 16, 2024.
Tarakcioglu, Emre. "Melatonin on NLRP3 Inflammasome Activation" Encyclopedia, https://encyclopedia.pub/entry/11726 (accessed April 16, 2024).
Tarakcioglu, E. (2021, July 06). Melatonin on NLRP3 Inflammasome Activation. In Encyclopedia. https://encyclopedia.pub/entry/11726
Tarakcioglu, Emre. "Melatonin on NLRP3 Inflammasome Activation." Encyclopedia. Web. 06 July, 2021.
Melatonin on NLRP3 Inflammasome Activation
Edit

The NLRP3 inflammasome is a part of the innate immune system and responsible for the rapid identification and eradication of pathogenic microbes, metabolic stress products, reactive oxygen species, and other exogenous agents. NLRP3 inflammasome is overactivated in several neurodegenerative, cardiac, pulmonary, and metabolic diseases. Therefore, suppression of inflammasome activation is of utmost clinical importance. Melatonin is a ubiquitous hormone mainly produced in the pineal gland with circadian rhythm regulatory, antioxidant, and immunomodulatory functions. Melatonin is a natural product and safer than most chemicals to use for medicinal purposes. Many in vitro and in vivo studies have proved that melatonin alleviates NLRP3 inflammasome activity via various intracellular signaling pathways.

melatonin NLRP3 inflammasome disease inflammation

1. Introduction

The NLRP3 inflammasome is a crucial step in innate immune responses and contributes to immune pathogenesis of several diseases including neurodegenerative, cardiac, pulmonary, gastrointestinal, and metabolic diseases. Therefore, inhibition of NLRP3 inflammasome activation is a novel therapeutic target for inflammation-related disorders. In recent years, many natural or synthetic molecules targeting NLRP3 have been evaluated for this purpose. Melatonin is one of the best candidate agents due to it being a natural and endogenous molecule. Figure 1 proposes melatonin's essential protective actions on the inflammatory pathways associated with NLRP3 inflammasome activation, which are reviewed in this entry. By virtue of these protective mechanisms, melatonin is a promising molecule for the treatment of inflammation-related disorders.
Figure 1. Representation of melatonin effects on NLRP3 inflammasome activation. Melatonin shows ameliorative effects on NLRP3 inflammasome activation via interacting with certain signaling pathways. NLRP3: NLR family pyrin domain containing 3; ASC: apoptotic-associated speck-like protein containing a caspase recruitment domain; ROS: reactive oxygen species; TXNIP: thioredoxin-interacting protein; Nrf2: nuclear factor erythroid 2-related factor 2; Sirt-1: sirtuin 1, IL-1β: interleukin 1β; NF-κB: nuclear factor kappa B.

2. Melatonin

Melatonin (N-acetyl-5-methoxy tryptamine) is a hormone produced from L-tryptophan via a chain of enzymatic reactions, mostly in the pineal gland [1][2]. One of the well-recognized functions of melatonin is circadian rhythm regulation. Additionally, anti-inflammatory, cytoprotective, free radical scavenging, antioxidative, anti-cancer, anti-aging, and immunomodulatory effects are also reported [3][4][5]. Two different membrane receptors for melatonin binding have been identified until today: Melatonin may bind to high-affinity MT1 (Mel1a) and MT2 (Mel1b) receptors which belong to the G protein-coupled receptor (GPCR) family [6]. Other than cell membrane receptors, melatonin can bind to the intracellular MT3 (Mel1c) receptor [7], calmodulin [8], calreticulin [9], and retinoic acid receptor superfamily (retinoid Z receptors/retinoid orphan receptors (RZR/ROR)): RORα splicing variants (RORα1, RORα2, RORα3, RZRα), RZRβ, and RORγ [10][11]. In addition to interactions with all receptors, melatonin has the ability to pass through membranes due to its amphiphilicity; so, it may react with molecules in cells without the help of receptors, termed as non-receptor mediated actions [12]. A complete overview of anti-inflammatory actions of melatonin can be found in articles published previously [13][14]. In this entry, we focus on the NLR family pyrin domain containing 3 (NLRP3) inflammasome, and we summarize studies on the effects of melatonin on NLRP3 inflammasome activation in a variety of diseases and possible intracellular mechanisms of these effects.

3. NLRP3 Inflammasome and Regulation

The inflammatory response is a crucial response for survival, governed by pro-inflammatory cytokines and chemokines. The innate immune system is the first to encounter pathogens and threats to the organism and respond immediately. Hyperactivation or abnormal activation of the inflammation response leads to the high concentration of cytotoxic molecules, in turn leading to tissue damage [15]. Inflammasomes are vital contributors to innate immunity. These are cytosolic multiprotein complexes composed of a sensor protein (an AIM-like receptor or NOD-like receptor (NLR)), an adaptor protein, and an effector protein. When the inflammasomes are activated with a danger signal, cytokine secretion is triggered to clear pathogens or damaged cells [16]. The NLR family of cytosolic pattern recognition receptors are vital in recognizing intracellular bacterial breakdown products and starting an innate immunity cascade. It has several members such as NLRP1, NLRP2, NLRP3, NLRC4, NLRP7, NLRP6, and NLRP9b [17][18][19][20][21][22]. NLR family receptors contain a C-terminal leucine-rich repeat ligand sensing region (LRR), a NOD domain, and an N-terminal signaling module. This module may be a caspase recruitment domain (CARD), a pyrin domain (PYD), or a baculovirus inhibitor of apoptosis repeat [16].
The most extensively studied inflammasome complex is NLRP3, and it has a vast repertoire of recognition, enabling easy activation of immunity and pathogen clearance. The complex formation is activated via internal and/or external factors; the primary factors being pathogen-associated molecular patterns (PAMPs), danger-associated molecular patterns (DAMPs), reactive oxygen species (ROS), ion fluxes, lysosomal destabilization, ATP, and peptide aggregates [23]. Once activated on its LRR domain, NLRP3 recruits an adaptor protein called ASC (apoptosis-associated speck-like protein containing a CARD) on its pyrin domain. ASC oligomerizes with other recruited ASCs to form what is termed ASC specks, and in return, recruits pro-caspase-1 on its CARD containing end and triggers caspase-1 activation [23][24][25]. Pro-caspase-1 is an auto-proteolytic enzyme, and the proximity induced by ASC specks thus generates caspase-1. After activation, the complex triggers inflammatory cytokine secretion which ultimately leads to pyroptosis [26]. Canonical activation of the NLRP3 inflammasome requires two consecutive signals: priming and activation. The priming signal is initiated via TLR receptors by PAMPs such as LPS and leads to transcription of inflammatory cytokine genes through activation and nuclear translocation of NF-κB. The activation signal is the stimulation of NLRP3 by the various factors it recognizes, which eventually results in pro-caspase-1 cleavage [23]. Cleavage of pro-caspase-1 results in two enzymatically active caspase-1 subunits: p10 and p20 [27]. Active caspase-1 cleaves pro-inflammatory cytokines IL-1β, IL-18, and the protein Gasdermin D into their mature forms. Mature Gasdermin D forms pores on the cellular membrane, resulting in pyroptotic cell death and release of cytokines [28]. IL-1β and IL-18 amplify NLR-mediated inflammation and facilitate infiltration of immune cells [15].
Strict regulation of NLRP3 inflammasome is crucial. Post-transcriptional, post-translational, and negative regulation mechanisms enable tightly controlled NLRP3 activation [16]. At the post-translational level, the deubiquitinating enzyme BRCA1/BRCA2-containing complex subunit 3 (BRCC3) instigates NLRP3 activation [29]. On the other hand, F-box L2 [30], TRIM31 [31], and MARCH7 [32] were shown to alleviate NLRP3 inflammasome activation by ubiquitination of NLRP3. Protein tyrosine phosphatase non-receptor 22 [33] and protein phosphatase 2A [34] augment NLRP3 inflammasome by dephosphorylation, while Jun N-terminal kinase [35] and protein kinase D [36] augment NLRP3 inflammasome by phosphorylation of NLRP3.
Thioredoxin-interacting protein (TXNIP) regulates intracellular redox balance by inhibiting important antioxidant proteins called thioredoxins and, upon amplification of intracellular ROS levels, TXNIP dissociates from thioredoxins and induces NLRP3 activation by binding to it [37]. Endoplasmic reticulum (ER) stress is another known inducer of NLRP3 activation: induction of ER stress and the unfolded protein response results in TXNIP upregulation [38][39] and Ca2+ release into the cytosol, which can damage mitochondria and lead to ROS accumulation [40]. Mitochondrial damage can also release other stimulators of NLRP3 into the cytosol: cardiolipin and oxidized mtDNA [41]. One more organelle that is implicated in NLRP3 activity is the lysosome: damage of lysosomes is known to release cathepsin B into the cytosol, where it induces NLRP3 activation [42][43]. Linking these together is autophagy, which can remove damaged organelles and thus relieve the cell of inflammatory signals. Indeed, autophagy has been reported multiple times as a negative regulator of NLRP3 [44]. At the post-transcriptional level, numerous miRNAs have been demonstrated to regulate NLRP3 inflammasome; miR-223 [45], miR-7 [46], miR-1929-3p [47] restrains NLRP3 inflammasome. Additionally, extensive studies indicate lncRNAs also have post-transcriptional regulatory effects on NLRP3 inflammasome activation; examples include nuclear enriched abundant transcript 1 (Neat1) [48], MIAT [49], Gm15441 [50], and Platr4 [51]. Certain molecules may bind to compounds in the NLRP3 inflammasome complex or other related molecules to inhibit it and provide negative regulation. Several proved negative regulators are B-cell adapter for phosphoinositide 3-kinase [52], PYRIN domain-only protein 1 [53] and 2 [54], TRIM30 [55], heat shock protein 70 [56], NLR family CARD-containing 3 protein [57] and autophagy [58].

4. The Mechanisms of the Action of Melatonin on NLRP3 Inflammasome Inhibition

Melatonin exerts inhibitory function on NLRP3 inflammasome activation through inhibiting or activating several proteins and pathways. NF-κB is a master regulator of the priming phase of NLRP3 inflammasome activation. Melatonin prevents NLRP3 inflammasome activation by inhibiting NF-κB signaling via RORα [59] and silent information regulator 1 (SIRT1)-dependent deacetylation of NF-κB [59]. ROS is a main trigger of NLRP3 inflammasome activation. Growing evidence shows that melatonin reduces levels of TXNIP, leading to suppression of ROS production and NLRP3 activity [60][61]. TXNIP is also mediator of ER stress induced NLRP3 inflammasome activation. Melatonin downregulates ER-induced TXNIP/NLRP3 pathway in LPS-induced endometritis [61]. SIRT1 is a NAD+ dependent deacetylase and a strong regulator of inflammatory, metabolic, and oxidative stressors of cells [62]. Preceding studies reported that sirtuins attenuate NLRP3 inflammasome activation by deacetylating of NLRP3 protein [63], and melatonin increased SIRT1 activity to inhibit the NLRP3 inflammasome [62][64]. Another pathway that melatonin may modulate is Nrf2 which is an antioxidant protein promoting ROS clean-up [17]. It has been documented that melatonin displays protection against NLRP3 inflammasome activity through Nrf2-mediated ROS scavenging and elimination [64][65]. It is well known that autophagy is a negative regulator for NLRP3 activation. Mitophagy is a subtype of autophagy that helps elimination of dysfunctional mitochondria. Melatonin increased the expression of LC3-II/LC3-I and Atg 5 as autophagy markers and Parkin and PINK-1 as mitophagy markers while suppressing NLRP3 inflammasome in the Subarachnoid hemorrhage (SAH) model [59]. Since 3-MA, an autophagy inhibitor, reverses these beneficial effects of melatonin on NLRP3 inflammasome, the results suggest that the effect of melatonin on NLRP3 inflammasome inhibition is dependent on mitophagy induction. The regulatory function of melatonin on inflammasome partly occurs through post-transcriptional mechanisms. Melatonin inhibits NLRP3 inflammasome complex formation by altering the expression of miRNAs and long noncoding RNAs [66][67][68]. Melatonin was tested against pyroptotic cell death in endothelial cells and, it reduced pyroptotic cell death via noncoding RNA MEG3/miR-223/NLRP3 [69]. Furthermore, melatonin mitigated cardiac fibrosis in mice by blocking lncRNA MALAT1/miR-141-mediated NLRP3 inflammasome activation [66]. Melatonin healed radiation induced lung injury both in vivo and in vitro by suppressing miR-30e/NLRP3 axis [68].
The NLRP3 inflammasome is known to have a critical role in the pathogenesis of many diseases and conditions with inflammatory components [70][71][72][73]; and the suppressive effect of melatonin on the NLRP3 inflammasome has been demonstrated in various in vitro and in vivo models of diseases and injuries. The detailed review of melatonin's reported effects on NLRP3 in these diseases can be found in the full review article associated with this entry.

References

  1. Song, R.; Ren, L.; Ma, H.; Hu, R.; Gao, H.; Wang, L.; Chen, X.; Zhao, Z.; Liu, J. Melatonin promotes diabetic wound healing in vitro by regulating keratinocyte activity. Am. J. Transl. Res. 2016, 8, 4682–4693.
  2. Claustrat, B.; Leston, J. Melatonin: Physiological effects in humans. Neurochirurgie 2015, 61, 77–84.
  3. Bhattacharya, S.; Patel, K.K.; Dehari, D.; Agrawal, A.K.; Singh, S. Melatonin and its ubiquitous anticancer effects. Mol. Cell. Biochem. 2019, 462, 133–155.
  4. Cardinali, D.P. Melatonin: Clinical Perspectives in Neurodegeneration. Front. Endocrinol. 2019, 10, 480.
  5. Meng, X.; Li, Y.; Li, S.; Zhou, Y.; Gan, R.Y.; Xu, D.P.; Li, H.B. Dietary Sources and Bioactivities of Melatonin. Nutrients 2017, 9, 367.
  6. Hardeland, R. Melatonin: Signaling mechanisms of a pleiotropic agent. Biofactors 2009, 35, 183–192.
  7. Nosjean, O.; Ferro, M.; Coge, F.; Beauverger, P.; Henlin, J.M.; Lefoulon, F.; Fauchere, J.L.; Delagrange, P.; Canet, E.; Boutin, J.A. Identification of the melatonin-binding site MT3 as the quinone reductase 2. J. Biol. Chem. 2000, 275, 31311–31317.
  8. Benitez-King, G.; Anton-Tay, F. Calmodulin mediates melatonin cytoskeletal effects. Experientia 1993, 49, 635–641.
  9. Macias, M.; Escames, G.; Leon, J.; Coto, A.; Sbihi, Y.; Osuna, A.; Acuna-Castroviejo, D. Calreticulin-melatonin. An unexpected relationship. Eur. J. Biochem. 2003, 270, 832–840.
  10. Becker-Andre, M.; Wiesenberg, I.; Schaeren-Wiemers, N.; Andre, E.; Missbach, M.; Saurat, J.H.; Carlberg, C. Pineal gland hormone melatonin binds and activates an orphan of the nuclear receptor superfamily. J. Biol. Chem. 1994, 269, 28531–28534.
  11. Smirnov, A.N. Nuclear melatonin receptors. Biochemistry 2001, 66, 19–26.
  12. Cipolla-Neto, J.; Amaral, F.G.D. Melatonin as a Hormone: New Physiological and Clinical Insights. Endocr. Rev. 2018, 39, 990–1028.
  13. Mauriz, J.L.; Collado, P.S.; Veneroso, C.; Reiter, R.J.; Gonzalez-Gallego, J. A review of the molecular aspects of melatonin’s anti-inflammatory actions: Recent insights and new perspectives. J. Pineal Res. 2013, 54, 1–14.
  14. Hardeland, R. Melatonin and inflammation-Story of a double-edged blade. J. Pineal Res. 2018, 65, e12525.
  15. Olcum, M.; Tastan, B.; Ercan, I.; Eltutan, I.B.; Genc, S. Inhibitory effects of phytochemicals on NLRP3 inflammasome activation: A review. Phytomedicine 2020, 75, 153238.
  16. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36.
  17. Janowski, A.M.; Sutterwala, F.S. Atypical Inflammasomes. Methods Mol. Biol. 2016, 1417, 45–62.
  18. Kerur, N.; Veettil, M.V.; Sharma-Walia, N.; Bottero, V.; Sadagopan, S.; Otageri, P.; Chandran, B. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe 2011, 9, 363–375.
  19. Khare, S.; Dorfleutner, A.; Bryan, N.B.; Yun, C.; Radian, A.D.; de Almeida, L.; Rojanasakul, Y.; Stehlik, C. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity 2012, 36, 464–476.
  20. Levy, M.; Thaiss, C.A.; Zeevi, D.; Dohnalova, L.; Zilberman-Schapira, G.; Mahdi, J.A.; David, E.; Savidor, A.; Korem, T.; Herzig, Y.; et al. Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell 2015, 163, 1428–1443.
  21. Minkiewicz, J.; de Rivero Vaccari, J.P.; Keane, R.W. Human astrocytes express a novel NLRP2 inflammasome. Glia 2013, 61, 1113–1121.
  22. Monroe, K.M.; Yang, Z.; Johnson, J.R.; Geng, X.; Doitsh, G.; Krogan, N.J.; Greene, W.C. IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science 2014, 343, 428–432.
  23. Jo, E.K.; Kim, J.K.; Shin, D.M.; Sasakawa, C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol. Immunol. 2016, 13, 148–159.
  24. de Zoete, M.R.; Palm, N.W.; Zhu, S.; Flavell, R.A. Inflammasomes. Cold Spring Harb. Perspect. Biol. 2014, 6, a016287.
  25. Voet, S.; Srinivasan, S.; Lamkanfi, M.; van Loo, G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol. Med. 2019, 11.
  26. Sharma, D.; Kanneganti, T.D. The cell biology of inflammasomes: Mechanisms of inflammasome activation and regulation. J. Cell. Biol. 2016, 213, 617–629.
  27. Alnemri, E.S.; Fernandes-Alnemri, T.; Litwack, G. Cloning and expression of four novel isoforms of human interleukin-1 beta converting enzyme with different apoptotic activities. J. Biol. Chem. 1995, 270, 4312–4317.
  28. de Vasconcelos, N.M.; Van Opdenbosch, N.; Van Gorp, H.; Parthoens, E.; Lamkanfi, M. Single-cell analysis of pyroptosis dynamics reveals conserved GSDMD-mediated subcellular events that precede plasma membrane rupture. Cell Death Differ. 2018.
  29. Py, B.F.; Kim, M.S.; Vakifahmetoglu-Norberg, H.; Yuan, J. Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol. Cell 2013, 49, 331–338.
  30. Han, S.; Lear, T.B.; Jerome, J.A.; Rajbhandari, S.; Snavely, C.A.; Gulick, D.L.; Gibson, K.F.; Zou, C.; Chen, B.B.; Mallampalli, R.K. Lipopolysaccharide Primes the NALP3 Inflammasome by Inhibiting Its Ubiquitination and Degradation Mediated by the SCFFBXL2 E3 Ligase. J. Biol. Chem. 2015, 290, 18124–18133.
  31. Song, H.; Liu, B.; Huai, W.; Yu, Z.; Wang, W.; Zhao, J.; Han, L.; Jiang, G.; Zhang, L.; Gao, C.; et al. The E3 ubiquitin ligase TRIM31 attenuates NLRP3 inflammasome activation by promoting proteasomal degradation of NLRP3. Nat. Commun. 2016, 7, 13727.
  32. Yan, Y.; Jiang, W.; Liu, L.; Wang, X.; Ding, C.; Tian, Z.; Zhou, R. Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell 2015, 160, 62–73.
  33. Spalinger, M.R.; Kasper, S.; Gottier, C.; Lang, S.; Atrott, K.; Vavricka, S.R.; Scharl, S.; Raselli, T.; Frey-Wagner, I.; Gutte, P.M.; et al. NLRP3 tyrosine phosphorylation is controlled by protein tyrosine phosphatase PTPN22. J. Clin. Investig. 2016, 126, 1783–1800.
  34. Stutz, A.; Kolbe, C.C.; Stahl, R.; Horvath, G.L.; Franklin, B.S.; van Ray, O.; Brinkschulte, R.; Geyer, M.; Meissner, F.; Latz, E. NLRP3 inflammasome assembly is regulated by phosphorylation of the pyrin domain. J. Exp. Med. 2017, 214, 1725–1736.
  35. Song, N.; Liu, Z.S.; Xue, W.; Bai, Z.F.; Wang, Q.Y.; Dai, J.; Liu, X.; Huang, Y.J.; Cai, H.; Zhan, X.Y.; et al. NLRP3 Phosphorylation Is an Essential Priming Event for Inflammasome Activation. Mol. Cell 2017, 68, 185–197.e186.
  36. Zhang, Z.; Meszaros, G.; He, W.T.; Xu, Y.; de Fatima Magliarelli, H.; Mailly, L.; Mihlan, M.; Liu, Y.; Puig Gamez, M.; Goginashvili, A.; et al. Protein kinase D at the Golgi controls NLRP3 inflammasome activation. J. Exp. Med. 2017, 214, 2671–2693.
  37. Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 2010, 11, 136–140.
  38. Oslowski, C.M.; Hara, T.; O’Sullivan-Murphy, B.; Kanekura, K.; Lu, S.; Hara, M.; Ishigaki, S.; Zhu, L.J.; Hayashi, E.; Hui, S.T.; et al. Thioredoxin-interacting protein mediates ER stress-induced beta cell death through initiation of the inflammasome. Cell Metab. 2012, 16, 265–273.
  39. Lerner, A.G.; Upton, J.P.; Praveen, P.V.; Ghosh, R.; Nakagawa, Y.; Igbaria, A.; Shen, S.; Nguyen, V.; Backes, B.J.; Heiman, M.; et al. IRE1alpha induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab. 2012, 16, 250–264.
  40. Cao, S.S.; Kaufman, R.J. Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxid. Redox Signal. 2014, 21, 396–413.
  41. Elliott, E.I.; Sutterwala, F.S. Initiation and perpetuation of NLRP3 inflammasome activation and assembly. Immunol. Rev. 2015, 265, 35–52.
  42. Amaral, E.P.; Riteau, N.; Moayeri, M.; Maier, N.; Mayer-Barber, K.D.; Pereira, R.M.; Lage, S.L.; Kubler, A.; Bishai, W.R.; D’Imperio-Lima, M.R.; et al. Lysosomal Cathepsin Release Is Required for NLRP3-Inflammasome Activation by Mycobacterium tuberculosis in Infected Macrophages. Front. Immunol. 2018, 9, 1427.
  43. 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, 128.
  44. Biasizzo, M.; Kopitar-Jerala, N. Interplay Between NLRP3 Inflammasome and Autophagy. Front. Immunol. 2020, 11, 591803.
  45. Bauernfeind, F.; Rieger, A.; Schildberg, F.A.; Knolle, P.A.; Schmid-Burgk, J.L.; Hornung, V. NLRP3 inflammasome activity is negatively controlled by miR-223. J. Immunol. 2012, 189, 4175–4181.
  46. Zhou, Y.; Lu, M.; Du, R.H.; Qiao, C.; Jiang, C.Y.; Zhang, K.Z.; Ding, J.H.; Hu, G. MicroRNA-7 targets Nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson’s disease. Mol. Neurodegener. 2016, 11, 28.
  47. Wang, Y.; Huang, Z.; Zhong, H.; Wang, L.; Xi, D.; Shi, Y.; Zhou, W.; Liu, Y.; Tang, N.; He, F. miR-1929-3p Overexpression Alleviates Murine Cytomegalovirus-Induced Hypertensive Myocardial Remodeling by Suppressing Ednra/NLRP3 Inflammasome Activation. BioMed Res. Int. 2020, 2020, 6653819.
  48. Dai, W.; Wang, M.; Wang, P.; Wen, J.; Wang, J.; Cha, S.; Xiao, X.; He, Y.; Shu, R.; Bai, D. lncRNA NEAT1 ameliorates LPSinduced inflammation in MG63 cells by activating autophagy and suppressing the NLRP3 inflammasome. Int. J. Mol. Med. 2020.
  49. Wang, Z.; Kun, Y.; Lei, Z.; Dawei, W.; Lin, P.; Jibo, W. LncRNA MIAT downregulates IL-1beta, TNF-a to suppress macrophage inflammation but is suppressed by ATP-induced NLRP3 inflammasome activation. Cell Cycle 2021, 1–10.
  50. Brocker, C.N.; Kim, D.; Melia, T.; Karri, K.; Velenosi, T.J.; Takahashi, S.; Aibara, D.; Bonzo, J.A.; Levi, M.; Waxman, D.J.; et al. Long non-coding RNA Gm15441 attenuates hepatic inflammasome activation in response to PPARA agonism and fasting. Nat. Commun. 2020, 11, 5847.
  51. Lin, Y.; Wang, S.; Gao, L.; Zhou, Z.; Yang, Z.; Lin, J.; Ren, S.; Xing, H.; Wu, B. Oscillating lncRNA Platr4 regulates NLRP3 inflammasome to ameliorate nonalcoholic steatohepatitis in mice. Theranostics 2021, 11, 426–444.
  52. Carpentier, S.J.; Ni, M.; Duggan, J.M.; James, R.G.; Cookson, B.T.; Hamerman, J.A. The signaling adaptor BCAP inhibits NLRP3 and NLRC4 inflammasome activation in macrophages through interactions with Flightless-1. Sci. Signal. 2019, 12.
  53. de Almeida, L.; Khare, S.; Misharin, A.V.; Patel, R.; Ratsimandresy, R.A.; Wallin, M.C.; Perlman, H.; Greaves, D.R.; Hoffman, H.M.; Dorfleutner, A.; et al. The PYRIN Domain-only Protein POP1 Inhibits Inflammasome Assembly and Ameliorates Inflammatory Disease. Immunity 2015, 43, 264–276.
  54. Ratsimandresy, R.A.; Chu, L.H.; Khare, S.; de Almeida, L.; Gangopadhyay, A.; Indramohan, M.; Misharin, A.V.; Greaves, D.R.; Perlman, H.; Dorfleutner, A.; et al. The PYRIN domain-only protein POP2 inhibits inflammasome priming and activation. Nat. Commun. 2017, 8, 15556.
  55. Hu, Y.; Mao, K.; Zeng, Y.; Chen, S.; Tao, Z.; Yang, C.; Sun, S.; Wu, X.; Meng, G.; Sun, B. Tripartite-motif protein 30 negatively regulates NLRP3 inflammasome activation by modulating reactive oxygen species production. J. Immunol. 2010, 185, 7699–7705.
  56. Martine, P.; Chevriaux, A.; Derangere, V.; Apetoh, L.; Garrido, C.; Ghiringhelli, F.; Rebe, C. HSP70 is a negative regulator of NLRP3 inflammasome activation. Cell Death Dis. 2019, 10, 256.
  57. Eren, E.; Berber, M.; Ozoren, N. NLRC3 protein inhibits inflammation by disrupting NALP3 inflammasome assembly via competition with the adaptor protein ASC for pro-caspase-1 binding. J. Biol. Chem. 2017, 292, 12691–12701.
  58. Yuan, X.; Bhat, O.M.; Meng, N.; Lohner, H.; Li, P.L. Protective Role of Autophagy in Nlrp3 Inflammasome Activation and Medial Thickening of Mouse Coronary Arteries. Am. J. Pathol. 2018, 188, 2948–2959.
  59. Garcia, J.A.; Volt, H.; Venegas, C.; Doerrier, C.; Escames, G.; Lopez, L.C.; Acuna-Castroviejo, D. Disruption of the NF-kappaB/NLRP3 connection by melatonin requires retinoid-related orphan receptor-alpha and blocks the septic response in mice. FASEB J. 2015, 29, 3863–3875.
  60. Cao, Z.; Fang, Y.; Lu, Y.; Tan, D.; Du, C.; Li, Y.; Ma, Q.; Yu, J.; Chen, M.; Zhou, C.; et al. Melatonin alleviates cadmium-induced liver injury by inhibiting the TXNIP-NLRP3 inflammasome. J. Pineal Res. 2017, 62.
  61. Hu, X.; Li, D.; Wang, J.; Guo, J.; Li, Y.; Cao, Y.; Zhang, N.; Fu, Y. Melatonin inhibits endoplasmic reticulum stress-associated TXNIP/NLRP3 inflammasome activation in lipopolysaccharide-induced endometritis in mice. Int. Immunopharmacol. 2018, 64, 101–109.
  62. Peng, Z.; Zhang, W.; Qiao, J.; He, B. Melatonin attenuates airway inflammation via SIRT1 dependent inhibition of NLRP3 inflammasome and IL-1beta in rats with COPD. Int. Immunopharmacol. 2018, 62, 23–28.
  63. He, M.; Chiang, H.H.; Luo, H.; Zheng, Z.; Qiao, Q.; Wang, L.; Tan, M.; Ohkubo, R.; Mu, W.C.; Zhao, S.; et al. An Acetylation Switch of the NLRP3 Inflammasome Regulates Aging-Associated Chronic Inflammation and Insulin Resistance. Cell Metab. 2020, 31, 580–591.e585.
  64. Arioz, B.I.; Tastan, B.; Tarakcioglu, E.; Tufekci, K.U.; Olcum, M.; Ersoy, N.; Bagriyanik, A.; Genc, K.; Genc, S. Melatonin Attenuates LPS-Induced Acute Depressive-Like Behaviors and Microglial NLRP3 Inflammasome Activation Through the SIRT1/Nrf2 Pathway. Front. Immunol. 2019, 10, 1511.
  65. Hou, Y.; Wang, Y.; He, Q.; Li, L.; Xie, H.; Zhao, Y.; Zhao, J. Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury. Behav. Brain Res. 2018, 336, 32–39.
  66. Che, H.; Wang, Y.; Li, H.; Li, Y.; Sahil, A.; Lv, J.; Liu, Y.; Yang, Z.; Dong, R.; Xue, H.; et al. Melatonin alleviates cardiac fibrosis via inhibiting lncRNA MALAT1/miR-141-mediated NLRP3 inflammasome and TGF-beta1/Smads signaling in diabetic cardiomyopathy. FASEB J. 2020, 34, 5282–5298.
  67. Che, H.; Li, H.; Li, Y.; Wang, Y.Q.; Yang, Z.Y.; Wang, R.L.; Wang, L.H. Melatonin exerts neuroprotective effects by inhibiting neuronal pyroptosis and autophagy in STZ-induced diabetic mice. FASEB J. 2020, 34, 14042–14054.
  68. Wu, X.; Ji, H.; Wang, Y.; Gu, C.; Gu, W.; Hu, L.; Zhu, L. Melatonin Alleviates Radiation-Induced Lung Injury via Regulation of miR-30e/NLRP3 Axis. Oxid. Med. Cell. Longev. 2019, 2019, 4087298.
  69. Zhang, Y.; Liu, X.; Bai, X.; Lin, Y.; Li, Z.; Fu, J.; Li, M.; Zhao, T.; Yang, H.; Xu, R.; et al. Melatonin prevents endothelial cell pyroptosis via regulation of long noncoding RNA MEG3/miR-223/NLRP3 axis. J. Pineal Res. 2018, 64.
  70. Xu, X.; Yin, D.; Ren, H.; Gao, W.; Li, F.; Sun, D.; Wu, Y.; Zhou, S.; Lyu, L.; Yang, M.; et al. Selective NLRP3 inflammasome inhibitor reduces neuroinflammation and improves long-term neurological outcomes in a murine model of traumatic brain injury. Neurobiol. Dis. 2018, 117, 15–27.
  71. Gris, D.; Ye, Z.; Iocca, H.A.; Wen, H.; Craven, R.R.; Gris, P.; Huang, M.; Schneider, M.; Miller, S.D.; Ting, J.P. NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses. J. Immunol. 2010, 185, 974–981.
  72. Halle, A.; Hornung, V.; Petzold, G.C.; Stewart, C.R.; Monks, B.G.; Reinheckel, T.; Fitzgerald, K.A.; Latz, E.; Moore, K.J.; Golenbock, D.T. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat. Immunol. 2008, 9, 857–865.
  73. Mangan, M.S.J.; Olhava, E.J.; Roush, W.R.; Seidel, H.M.; Glick, G.D.; Latz, E. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat. Rev. Drug Dis. 2018, 17, 588–606.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 461
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 07 Jul 2021
1000/1000