Next Article in Journal
Prenatal Molecular Hydrogen Administration Ameliorates Several Findings in Nitrofen-Induced Congenital Diaphragmatic Hernia
Previous Article in Journal
Genome-Wide Identification and Characterization of Short-Chain Dehydrogenase/Reductase (SDR) Gene Family in Medicago truncatula
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Use of Medicinal Plants and Their Bioactive Derivatives as Alveolar NLRP3 Inflammasome Regulators during Mycobacterium tuberculosis Infection

by
Nontobeko E. Mvubu
* and
Thamsanqa E. Chiliza
School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville 3630, South Africa
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9497; https://doi.org/10.3390/ijms22179497
Submission received: 30 June 2021 / Revised: 1 August 2021 / Accepted: 19 August 2021 / Published: 31 August 2021
(This article belongs to the Special Issue Innovative Approaches towards Mycobacterial Infections)

Abstract

:
Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), is a successful intracellular pathogen that is responsible for the highest mortality rate among diseases caused by bacterial infections. During early interaction with the host innate cells, M. tuberculosis cell surface antigens interact with Toll like receptor 4 (TLR4) to activate the nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3 (NLRP3) canonical, and non-canonical inflammasome pathways. NLRP3 inflammasome activation in the alveoli has been reported to contribute to the early inflammatory response that is needed for an effective anti-TB response through production of pro-inflammatory cytokines, including those of the Interleukin 1 (IL1) family. However, overstimulation of the alveolar NLRP3 inflammasomes can induce excessive inflammation that is pathological to the host. Several studies have explored the use of medicinal plants and/or their active derivatives to inhibit excessive stimulation of the inflammasomes and its associated factors, thus reducing immunopathological response in the host. This review describes the molecular mechanism of the NLRP3 inflammasome activation in the alveoli during M. tuberculosis infection. Furthermore, the mechanisms of inflammasome inhibition using medicinal plant and their derivatives will also be explored, thus offering a novel perspective on the alternative control strategies of M. tuberculosis-induced immunopathology.

Graphical Abstract

1. Introduction

Mycobacterium tuberculosis has proven to be a successful pathogen over the years, responsible for an estimated 1.2 million deaths, with 10 new million cases annually. This opportunistic pathogen is also accountable for another 0.2 million estimated deaths among HIV-TB co-infected individuals [1]. The success of M. tuberculosis as a human pathogen is due to the emergence, persistence and transmission of genetically diverse drug resistant strains in different geographic locations [2], delays in diagnosis [3,4], and failure of the individual’s immune system to contain and eliminate the pathogen [5].
In the initial stages of pulmonary TB infection, physical barriers and innate immune response are present to prevent and control the infection. Initial interaction between the M. tuberculosis bacillus and the alveolar lining present an opportunity for containment of infection by innate cells, such as type I and type II pulmonary epithelial cells [6], resident alveolar macrophages, and surrounding dendritic cells and neutrophils [7,8]. The highly abundant pulmonary epithelial cells in the alveolar lining are connected by tight junctions, adherens junctions, and desmosomes [9,10] that are relatively impermeable [11]. Residence alveolar macrophages are ideally located to render microbicidal action through the activation of phago-lysosome against invading bacilli [12]. Surrounding dendritic cells and neutrophils link the innate and adaptive immune responses and produce toxic mediators to eliminate the invading bacilli, respectively [13,14]. M. tuberculosis is a successful pathogen due to its ability to bypass the host innate defence mechanisms, which eventually leads to the granuloma formation [15].
Innate cells express a range of host pathogen recognition receptors (PRRs) that interact with pathogen associated molecular patterns (PAMPs) in M. tuberculosis [16,17,18]. Interaction of the M. tuberculosis cell surface lipid antigens with the host [18] innate cells receptors such as TLR4 stimulate the activation of the NLRP3 inflammasome pathway through the Myeloid differentiation primary response 88 (MyD88) adaptor molecule [19,20]. Activation of the NLRP3 inflammasome is critical in the production of inflammatory cytokines of the IL1 family [21,22]. Several studies have shown that inflammasome activation and early production of IL1 cytokine family contribute to the early immune response to M. tuberculosis, thus, offering an effective control strategy [23,24]. Paradoxically, overstimulation of the inflammasome resulting in high production of IL1 cytokines has been shown to be detrimental to the host due to overstimulation of the immune system, leading to immunopathology in the host [25,26]. Thus, controlling overstimulation of the alveolar inflammasome provides an ideal target for prevention of the inflammatory response that is associated with immunopathology during TB infection.
Several studies have shown the effectiveness of medicinal plants and their bioactive derivatives as a promising approach to TB treatment since these are naturally sourced compounds with minimal side effects [27,28,29,30]. Medicinal plants and their respective active compounds exhibit their anti-inflammatory mechanisms through inhibition of the NLRP3 inflammasome-associated transcripts [31,32], inflammasome assembly [33,34] and production of inflammasome proteins, including IL1 cytokines [35] in in vitro and in vivo M. tuberculosis infection models. The current review reveals the molecular mechanism behind inflammasome activation on resident alveolar macrophages and pulmonary epithelial cells that results in IL1 cytokine production, which have been shown to contribute to the alveolar inflammatory response during M. tuberculosis infection. Furthermore, medicinal plants and their active derivatives that can be explored to prevent overstimulation of the alveolar NLRP3 inflammasome and their mechanisms of action will be reported as a novel host-directed therapy against M. tuberculosis.

2. M. tuberculosis Interaction with Alveolar Macrophages and Pulmonary Epithelial Cells

Initial interaction between M. tuberculosis PAMPs and mammalian PRRs is crucial for the bacilli to gain entry into the cells and initiate the pathogenesis process. Several studies have shown that macrophages express complement (CR) [36,37] and Fc gamma (Fcγ) receptors [38] during M. tuberculosis infection. Reduced M. tuberculosis bacterial uptake was observed in macrophages isolated from CR3-knockout mice compared to the wild type [39]. Moreover, several studies [40,41,42] have revealed an important role of CR receptors in opsonic and non-opsonic phagocytosis of M. tuberculosis in human macrophages. Initially, the Fcγ receptors were thought to contribute to M. tuberculosis killing because they mediate uptake of Immunoglobulin G (IgG)-opsonised mycobacteria [43], however, they were later shown to reduce the Th1 response by attenuating IL-12p40 production [38]. Collectively, these findings suggest that both CR and Fcγ receptors are involved in M. tuberculosis uptake in macrophages through opsonic and non-opsonic phagocytosis.
Mannose receptors (MR) are transmembrane C-type (calcium dependent) lectins that recognise and bind to the expressed mannose sugars in the lipoarabinomannan (LAM) of M. tuberculosis surface [44,45,46,47]. Interaction between the MR and M. tuberculosis mannosylated LAM promotes phagocytosis in macrophages [44], which may be dependent on virulence of the strain [47]. Other well characterised receptors that contribute to the update of M. tuberculosis in alveolar macrophages include Surfactant Protein A [48,49], Dectin-1 [50,51], NOD2 [52], and CD14 [53]. TLRs [54,55] are one of the well-known host PRRs that are expressed by alveolar macrophages in recognition of the diverse PAMPs in M. tuberculosis surface as excellently reviewed by Faridgohar and Nikoueinejad [18]. Despite the advances and knowledge generated on host PRRs expressed on alveolar macrophages, differential activation of these receptors by different lineages of M. tuberculosis remains to be investigated as this can contribute to better understanding of the inflammatory response induced by genetically diverse clinical strains.
Previously, not much was known about specific receptors expressed by pulmonary epithelial cells during M. tuberculosis infection. Bermudez and Goodman [56] initially proposed that M. tuberculosis use microtubules and microfilament pathways to gain entry into pulmonary epithelial cells. This was shown by significant reduction of intracellular bacilli when these pathways were blocked [56]. Lee et al. [57] revealed an upregulation of Dectin-1 receptors in a TLR-2 dependent manner of A549 pulmonary epithelial cells during M. tuberculosis infection. Through the production of a Dectin-1 receptor protein, they showed that these non-phagocytic cells might use this receptor for recognition of M. tuberculosis. The M. tuberculosis strains of East-African Indian and Euro-American lineages both induced down-regulation of the NOD1 receptor of the A549 pulmonary epithelial cells at 72 h post-infection [58]. However, a time course analysis of the NOD-associated receptors is needed to establish their involvement in the recognition of M. tuberculosis. Mannose and DC-SIGN receptors have not been linked to M. tuberculosis invasion of pulmonary epithelial cells and this area remains to be investigated.
TLRs are involved in the recognition of different M. tuberculosis cell wall-associated structures, such as acylated lipoproteins (TLR1, TLR2, TLR6), ESAT-6, mycolic acid (TLR2), acylated lipomannan (TLR4), and trehalose dimycolates (TLR3, TLR4, and TLR9), respectively [18]. Sequeira et al. [59] showed the role of TLR2 of pulmonary epithelial cells in the production of pro-inflammatory cytokines during infection by M. tuberculosis. Inhibition of TLR2-mediated response by mycolic acid and mce1 operon mutant significantly reduce the production of both IL8 and MCP-1, which suggests that TLR2 is essential to effective inflammatory response of pulmonary epithelial cells to M. tuberculosis. Previously, we showed an increased expression of TLR2, TLR3, TLR5, and TLR8 in pulmonary epithelial cells at 48 h post-infection by genetically diverse M. tuberculosis clinical strains of East-Asian and Euro-American lineages, leading to production of a diverse range of inflammatory cytokines. Upregulation of TLR4 was only induced by the Beijing (East-Asian) and F11 (Euro-American) strains, while TLR1, TLR6, TLR7, and TLR9 were downregulated by all strains [60].
A transcriptomic study performed by Hadifa et al. [58] revealed an increased expression of TLR1 and TLR3 by East-African Indian and Euro-American lineages of M. tuberculosis at 72 h post-infection of A549 pulmonary epithelial cells. Furthermore, the East-African Indian strain induced increased expression of TLR2, TLR5, and TLR9; while the Euro-American strain induced down-regulation of TLR4, TLR5, and TLR9. Both transcriptomic studies [58,60] indicate the TLRs are activated by different lineages of M. tuberculosis in pulmonary epithelial cells at 48 and 72 h post infection and these PRRs may be the main receptors used by this pathogen to gain entry into these cells.
Upon stimulation of the alveolar macrophages and pulmonary epithelial cells TLRs by M. tuberculosis PAMPs, the MyD88, TIR-domain-containing adapter-inducing interferon-β (TRIF), TRIF-related adapter molecule (TRAM), Toll-interleukin 1 receptor domain containing adaptor protein (TIRAP), B-cell adaptor for PI3K (BCAP), and sterile alpha and TIR motif containing (SARM) are adaptor molecules that are central components of the TLR signalling pathway [61]. Previously, we showed that genetically diverse strains of M. tuberculosis induced high enrichment of the TLR signalling pathways in pulmonary epithelial cells with increased expression of the MyD88 adaptor molecule [62]. Several studies have linked phagocytosis of M. tuberculosis by alveolar macrophages to the MyD88-signalling pathway [63,64] with a highly impaired inflammatory response in MyD88-knockout studies [65,66]. Thus, the activation of these adaptor molecules in pulmonary epithelial cells and alveolar macrophages results in downstream signalling cascade, leading to the activation transcriptional factors that are responsible for expression of inflammatory cytokines, including interferons and IL1 cytokine family [67]. Moreover, interaction between M. tuberculosis surface antigens with TLR4 triggers the NF-kB signalling pathway to induce expression of IL1β and IL18, whose maturation and production is dependent on the inflammasome assembly [68].

3. Activation of the Alveolar NLRP3 Inflammasome during M. tuberculosis Infection

Inflammasome is a multi-protein complex intracellular structure, which induces maturation of inflammatory cytokines, IL1β, and IL18 through the activation of caspase-1 and caspase-4/11 [69]. The activation of the inflammasome is triggered by signalling pathways that involves host PRRs, such as NOD-like receptors and TLRs [19]. Inflammasomes function in modulating host defence response, as well as pyroptosis, which is an inflammatory induced lytic mode of cell death that can be caspase-1 or caspase-11 dependent [70]. The assembly of an inflammasome is a coordinated signalling event, which is essential to producing an immune response after sensing M. tuberculosis [71,72]. The canonical NLRP3 inflammasomes is mediated by the activation of Caspase 1, while the non-canonical NLRP3 activation can be activated downstream of Caspase 4/11, respectively [19,67].
The canonical inflammasome pathway is initiated through the activation of caspase 1 by NLRP3, which responds to stimulation by IL1R, Tumor necrosis factor (TNF) or a TLR ligand that binds its cognate receptor, which results in the translocation of NF-κB into the nucleus. Thereafter, the expression of NLRP3 and pro-IL1β are induced, while pro-IL18 is constitutively expressed within cells. Several studies [18,54,55,58,60] have shown that M. tuberculosis activate TLR4 that initiate the canonical inflammasome in alveolar macrophages and pulmonary epithelial cells. Infection by genetically diverse clinical strains of M. tuberculosis revealed an upregulation of NFKB1 and NFKB2 transcriptional factors that lead to the transcription of IL1β, while IL18 was slightly down-regulated at 48 h infection by East-Asian and Euro-American lineages [60]. The Euro-American lineage strain induced upregulation of both NFKB1 and NFKB2 transcriptional factors at 72 h, while both transcripts were downregulated by the East-African Indian lineage strain, leading to downregulation of IL1β by both strains [58]. The M. tuberculosis Beijing strains of the East-Asian lineage were differentially recognised by TLR2 and TLR4 in macrophages resulting in distinct cytokine profiles, including IL1β [73], activating the canonical and non-canonical NF-κB signalling pathways [74]. Collectively, these findings indicate that M. tuberculosis interact with the TLR4, resulting in the activation of the NF-κB transcriptional factor, which is responsible for the transcription of pro-inflammatory cytokines, including IL1 by the alveolar macrophages and pulmonary epithelial cells. Furthermore, M. tuberculosis lineage-specific differences of the canonical inflammasome activation suggest that genetically diverse strains may induced diverse inflammasome patterns in the alveolar lining.
The second signal of the canonical inflammasome pathway involves the assembly of the NLRP3 inflammasome complex, which results in the recruitment of adaptor molecule, CARD and pro-caspase-1 in order to induce the processing and secretion of IL1β and IL18 cytokines. Pulmonary epithelial cells exhibit increased regulation of NLRP3 (by only F11 and H37Rv strains) PYCARD (ASC) and CASP1 during early infection by clinical strains of M. tuberculosis [60]. This upregulation may be time and lineage specific since the Euro-American lineage downregulated CASP1 at 72 h [58] in pulmonary epithelial cells. High levels of the mature IL18 were predominantly observed in M. tuberculosis-stimulated pulmonary epithelial cells, which indicated the upregulation of IL18 expression at both transcriptional and post-transcriptional levels, suggesting the involvement of CASP1 enzymatic activity [75]. There were no significant differences in the production of IL1β by pulmonary epithelial cells, which increased from 24, 48, and 72 h post-infection by wild type, Mycobacterium tuberculosis curli pili (MTP) mutant and complemented strains of M. tuberculosis [76], suggesting that IL1 cytokine production is not dependent on the MTP antigen. It should be noted that the activation of the canonical NLRP3 inflammasome pathway in pulmonary epithelial cells, if present, may be dependent on the lineage of the infecting strain, as well as the infection time as shown by the differences observed for the Euro-American and East-African Indian lineages [58,60]. Stimulation of the canonical inflammasome by Mycobacterium and it associated factors is well described and characterised in alveolar macrophages [72,77,78], as these cells have been shown to produce inflammatory IL1β and IL18 [77,78,79].
Non-canonical NLRP3 inflammasome mediated by CASP4/11 [80] has been shown to be activated by intestinal Gram-negative bacterial such as Citrobacter rodentium, Escherichia coli, Legionella pneumophila, Salmonella typhimurium, and Vibrio cholera [67] and other parasites such as Leishmania amazonensis [81]. The pathogen’s LPS and other surface antigens have been associated with non-canonical NLRP3 inflammasome activation through interaction with Guanylate binding proteins (GBPs). To our knowledge, there is no evidence of the non-canonical stimulation of the NLRP3 inflammasome by M. tuberculosis in pulmonary epithelial cells from transcriptomics and proteomics studies; this area remains to be investigated. However, the NLRP3 CASP4/11-dependant inflammasome is activated in macrophages during M. tuberculosis infection [82]. Collectively, these studies indicate that M. tuberculosis stimulate alveolar macrophages canonical and non-canonical NLRP3 inflammasome, leading to production of inflammatory cytokines, IL-1β, and IL18 in a TLR4-NF-kB dependent manner. Furthermore, despite increased expression of the NLRP3 inflammasome transcripts and subsequence IL1 cytokine production in pulmonary epithelial cells, the presence of this complex and other types of inflammasomes such as NLRP6, NLRC4, and AIM2 remains to be investigated and confirmed in future studies. The activation of a canonical and non-canonical alveolar NLRP3 molecular mechanism during M. tuberculosis infection is depicted in Figure 1.
It is apparent that alveolar macrophages produce IL1 cytokines [72,75,76,82,83] through the activation of canonical and/or non-canonical NLRP3 inflammasome by the TLR-MYD88 signalling pathway, which results in stimulation of NF-κB transcriptional factors that transcribe pro-inflammatory cytokines during M. tuberculosis infections. However, this mechanism remains to be confirmed for pulmonary epithelial cells. IL1 are among the group of pro-inflammatory cytokines that are essential in host defence against M. tuberculosis [77]. Mice with IL1α and IL1β double knockouts and IL1R type I-deficient mice display a defective granuloma phenotype accompanied with increased mycobacterial growth [79]. IL18 is known as an interferon-γ inducing factor, which has a vital role in T helper1 (Th1) response [84,85] and has been shown to be responsible for the production of pro-inflammatory cytokines, chemokines, and transcriptional factors [86,87]. Moreover, IL18 defective mice were shown to be highly susceptible to M. tuberculosis and BCG strains [88]. Despite the critical role played by IL1 cytokines of the NLRP3 inflammasome, overproduction of pro-inflammatory cytokines without efficient anti-inflammatory response has been associated with increase disease severity and high bacterial burden [89] that can compromise effective host response during TB infection. The presence of IL1β was associated with caseous granulomatous inflammation during M. tuberculosis, while blocking IL1β production relieved pulmonary inflammation [90]. Significantly high IL1β and IL18 was identified in drug resistant compared to drug susceptible TB [91], while high cytokine concentrations were identified in patients with a severe disease [92]. Therefore, the ability of pulmonary epithelial cells and alveolar macrophages to produce IL1 (IL1β and IL18) cytokines through the activation of the inflammasome may contribute to the inflammatory response during early infection by M. tuberculosis that is pathological to the host, thus novel strategies must be exploited to regulate this response.

4. Medicinal Plants and Their Bioactive Derivatives as Regulators of Alveolar NLRP3 Inflammasome during M. tuberculosis Infection

The high prevalence of TB worldwide is partly due to development of drug resistant strains, rendering the current treatment regimen ineffective [93,94,95]. Several studies have proposed the use of medicinal plants or their bioactive compounds with antimycobacterial activity [29,33,96,97,98,99]. However, recent studies have also exploited the use of medicinal plants and/or their derivatives that are regulators of the immune system during M. tuberculosis infection [100,101,102]. Medicinal plants and their respective bioactive compounds that regulate the components of the immune system may be a promising strategy in controlling Mycobacterial infections because they are not directly targeting the bacillus, reducing the pathogen’s need to develop drug resistance and selective pressure [103]. Therefore, medicinal plants and their bioactive derivatives may provide a novel host-directed perspective in controlling immunopathology in the alveoli through inhibition of the NLRP3 inflammasomes pathways that are crucial in the production of IL1β and IL18 cytokines during M. tuberculosis infections. Medicinal plants and/or their bioactive derivatives that are used to inhibit NLRP3 inflammasome gene transcription, formation of the inflammasome complex and production of NLRP3 inflammasome proteins (including IL1 cytokines) are reviewed below.

4.1. NLRP3 Inflammasome Transcript Inhibitors

Michelia compressa and Michelia champaca plants from the Magnolia family [104] possess the natural guaianolide sesquiterpene lactone, micheliolide, with a potent anti-inflammatory activity [105,106,107,108]. Zhang et al. [31] used an in vitro mouse macrophage-like cell line to investigate the anti-inflammatory activity of micheliolide during M. tuberculosis infection. The NF-κB transcriptional factor and NLRP3 inflammasome that are essential for transcription of IL1β, IL18, and pro-caspase 11 transcripts were downregulated, accompanied by reduction of inflammatory cytokines (IL1β and TNF-α) in a dose-dependent manner. This study concluded that the micheliolide downregulates the activation of NLRP3 inflammasome by modulating the inflammatory response induced by M. tuberculosis through the PI3K/Akt/NF-κB pathway.
NLRP3 inflammasome-specific transcript can be inhibited through the adaptor molecules that signal to activate transcriptional factor, NF-κB. Guttiferone K is the active compound isolated from Garcinia yunnanensis Hu plant [109] and has anti-cancer activities, including inhibition of autophagy and metastasis while promoting apoptosis of cancer [110]. The anti-inflammatory activity of Guttiferone K was observed through phosphorylation inhibition of the NF-κB transcriptional factor by Interleukin-1 receptor associated kinase (IRAK1) in the TLR signalling pathway of the M. tuberculosis H37Ra (avirulent) infected macrophages. This inhibition resulted in significantly reduced TLR-NF-κB inflammatory mediators, IL1β, TNF-α, IL6, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) [32]. The Guttiferone K anti-inflammatory molecular mechanism remains to be investigated in the virulent strains of M. tuberculosis.
There are many medicinal plant bioactive derivatives that act as NLRP3 inflammasome transcript inhibitors identified in non-M. tuberculosis models. These include Arctigenin [111], Silymarin [112], Rutin [113], Genistein [114], Aloe emodin [115], Anemoside B4 [116], epigallocathechin-3-gallate [117], Resveratrol [118], Silibinin [119], Artemisinin [120], Icariin [121], Polydatin [122], Cinnamaldehyde [123], Huangkui capsule [124] whose administration results in the inhibition of the inflammasome-associated transcription on both in vitro and in vivo models (Figure 2). It is crucial that these medicinal plant compounds be tested in M. tuberculosis in vitro and in vivo models to identify their potential use as host-directed immunomodulatory plant derivatives that can be used to prevent lung pathology during infection.

4.2. NLRP3 Inflammasome Protein Inhibitors

Medicinal plants and their derivatives can exert their anti-inflammatory activity in the NLRP3 inflammasome by directly inhibiting production of proteins that regulate this inflammatory pathway, including production of IL1 cytokines. The anti-inflammatory activity of andrographolide, the labdane diterpenoid isolated in the stems, and leaves of Andrographis paniculate was observed in macrophages co-cultured with pulmonary epithelial cells that had significantly reduced IL1β cytokine production during M. tuberculosis infection. This IL1β reduction in macrophages resulted in declining chemokine (IL8 and MCP-1) expression in neighbouring pulmonary epithelial cells. The proposed molecular mechanism behind IL1β inhibition is the activation of macrophage autophagy to degrade NLRP3, thus inhibiting the inflammasome activation and IL1β production. Moreover, andrographolide downregulated the phosphorylation of Akt/mTOR and NF-𝜅B p65 subunit, which will interfere with the NLRP3 inflammasome transcript expression that is essential for the maturation of the inflammasome complex [35]. Andrographolide is a natural labdane diterpenoid that was isolated in Andrographis paniculate plant [125] with many biological activities, including anti-cancer, antimicrobial, anti-diabetic, anti-inflammatory, etc. [126].
Several in vitro and in vivo models have identified other medicinal plant derivatives that have been found to inhibit NLRP3 inflammasome proteins and subsequent IL1 production; and these include Ginsenosides (Retinoblastoma and Compound K) [127,128], Curcumin [129], Genipin [130], Mangiferin [131], Salvianolate [132] that were derived from Panax ginseng, Curcuma longa, Gardenia jasminoides, Mangifera indica, Salvia miltiorrhiza medicinal plants, respectively.

4.3. Inhibitors of the NLRP3 Inflammasome Complex

Several medicinal plants and/or their bioactive derivatives can interfere with the formation of the inflammasome complex or the components that are needed for a complete NLRP3 inflammasome to facilitate maturation of the IL1 cytokines. The flavonoid Baicalin, isolated from Scutellaria baicalensis possess an anti-NLRP3 inflammasome activity during M. tuberculosis through decreased phosphorylated protein kinase B and mammalian target of rapamycin, decreasing NLRP3 inflammasome and subsequently IL1β production [34]. The COX-2 regulates the activation of the NLRP3 inflammasome through increased expression of LPS-induced pro-IL1β and NLRP3 by NF-κB [133]. The crude petroleum ether, ethanol, dichloromethane extracts of South African medicinal plants, Abrus precatorius subsp. Africanus, Ficus sur, Pentanisia prunelloides, and Terminalia phanerophlebia were shown to inhibit the COX-2 enzyme, thus may contribute to the anti-NLRP3 inflammasome response during M. tuberculosis infection [33]. The bioactive compound in these plants and their possible molecular mechanism against the NLRP3 inflammasome remains to be characterised.
NLRP3 inflammasome complex inhibitors in other models include Sweroside [134], Oridonin [135], Isoliquiritigenin [136], Cardamonin [137], Ginsenosides (Rg3, Rg1, 20S-protopanaxatriol, 25-OCH3-PPD) [138,139,140,141], Triptolide [142], Glycyrrhizin [143], Saikosaponin A [144], Quercetin [145], Dihydroquercetin [146], Casticin [147], Phloretin [148], Pterostilbene [149], Apocynin [150], Lycorine [151], Matrine [152], Tetramethylpyrazine [153], Astaxanthin [154], Danggui Buxue Tang [155], Quamoclit angulate plant [156], 4- Sulforaphane (methylsulfnylbutyl isothiocyanate) [157], Obovatol [158], Berberine [159] (Figure 2). Both in vitro and in vivo M. tuberculosis models exploring these bioactive plant derivatives remain to be investigated for their potential use as regulators of the alveolar NLRP3 inflammasome.
There is growing appreciation of medicinal plants and their bioactive molecules in management of inflammatory response regulated by the NLRP3 inflammasome as excellently reviewed by several authors [160,161,162,163]. To date, only five studies [31,32,33,34,35] have explored the use of medicinal plants and/or their bioactive derivatives in the management of the inflammasome in M. tuberculosis infection models. There is a growing understanding that M. tuberculosis infection is a multifaceted condition that is also driven by the host inflammatory response. Thus, controlling the alveolar NLRP3 inflammasome seem to be one of the novel strategies for host directed control measures of pathological response induced by M. tuberculosis.

5. Conclusions

The activation of the NLRP3 inflammasome in alveolar macrophages and potentially in pulmonary epithelial cells is an essential response of innate cells to M. tuberculosis that results in the production of IL1 inflammatory cytokines. Overstimulation of the alveolar NLRP3 inflammasome may contribute to the exaggerated inflammatory response that is pathological to the host. Recently, several studies have shown that medicinal plants and/or their derivatives, Andrographolide, Baicalin, Micheliolide, Guttiferone K, Abrus precatorius subsp. Africanus, Ficus sur, Pentanisia prunelloides, and Terminalia phanerophlebia can inhibit the NLRP3 inflammasome at transcriptional and post-transcriptional levels in M. tuberculosis models. Therefore, these plants and bioactive derivatives can be explored for anti-TB host directed adjuvants to control the M. tuberculosis-induced inflammatory response. Furthermore, many studies have identified many regulators of the NLRP3 inflammasome that remains to be tested on both in vitro and in vivo M. tuberculosis models. Future studies should investigate the ideal route of administration of these medicinal plants and their bioactive derivatives due to challenges associated with oral availability of medicinal drugs. Exploring alternative strategies such as using medicinal plants and their bioactive derivatives to regulate the host inflammatory response in TB may bring us closer to winning the battle against M. tuberculosis.

Author Contributions

Conceptualization, N.E.M.; writing—original draft preparation, N.E.M.; writing—review and editing, T.E.C.; visualization, N.E.M. and T.E.C.; funding acquisition, N.E.M. and T.E.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Research Foundation, Thuthuka grants, UID: 129635 and UID: 129846.

Acknowledgments

The authors would like to acknowledge Divenita Govender for administrative support in compiling the literature reviewed.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Global Tuberculosis Report 2019; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  2. Parwati, I.; van Crevel, R.; van Soolingen, D. Possible underlying mechanisms for successful emergence of the Mycobacterium tuberculosis Beijing genotype strains. Lancet Infect. Dis. 2010, 10, 103–111. [Google Scholar] [CrossRef]
  3. Getnet, F.; Demissie, M.; Assefa, N.; Mengistie, B.; Worku, A. Delay in diagnosis of pulmonary tuberculosis in low-and middle-income settings: Systematic review and meta-analysis. BMC Pulm. Med. 2017, 17, 202. [Google Scholar] [CrossRef] [PubMed]
  4. Bello, S.; Afolabi, R.F.; Ajayi, D.T.; Sharma, T.; Owoeye, D.O.; Oduyoye, O.; Jasanya, J. Empirical evidence of delays in diagnosis and treatment of pulmonary tuberculosis: Systematic review and meta-regression analysis. BMC Public Health 2019, 19, 820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bhat, K.H.; Yaseen, I. Mycobacterium tuberculosis: Macrophage takeover and modulation of innate effector responses. In Mycobacterium: Research and Development; InTech Open: London, UK, 2018. [Google Scholar]
  6. Ryndak, M.B.; Laal, S. Mycobacterium tuberculosis Primary Infection and Dissemination: A Critical Role for Alveolar Epithelial Cells. Front. Cell. Infect. Microbiol. 2019, 9, 299. [Google Scholar] [CrossRef] [PubMed]
  7. Whitsett, J.A.; Alenghat, T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat. Immunol. 2015, 16, 27–35. [Google Scholar] [CrossRef] [Green Version]
  8. Lerner, T.R.; Borel, S.; Gutierrez, M.G. The innate immune response in human tuberculosis. Cell Microbiol 2015, 17, 1277–1285. [Google Scholar] [CrossRef] [Green Version]
  9. Davies, J.A.; Garrod, D.R. Molecular aspects of the epithelial phenotype. Bioessays 1997, 19, 699–704. [Google Scholar] [CrossRef]
  10. Buckley, A.; Turner, J.R. Cell Biology of Tight Junction Barrier Regulation and Mucosal Disease. Cold Spring Harb. Perspect. Biol. 2018, 10, a029314. [Google Scholar] [CrossRef]
  11. Eisele, N.A.; Anderson, D.M. Host defense and the airway epithelium: Frontline responses that protect against bacterial invasion and pneumonia. J. Pathog. 2011, 2011, 249802. [Google Scholar] [CrossRef] [Green Version]
  12. Ehrt, S.; Schnappinger, D.; Bekiranov, S.; Drenkow, J.; Shi, S.; Gingeras, T.R.; Gaasterland, T.; Schoolnik, G.; Nathan, C. Reprogramming of the macrophage transcriptome in response to interferon-γ and Mycobacterium tuberculosis signaling roles of nitric oxide synthase-2 and phagocyte oxidase. J. Exp. Med. 2001, 194, 1123–1140. [Google Scholar] [CrossRef]
  13. Mihret, A. The role of dendritic cells in Mycobacterium tuberculosis infection. Virulence 2012, 3, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kroon, E.E.; Coussens, A.K.; Kinnear, C.; Orlova, M.; Möller, M.; Seeger, A.; Wilkinson, R.J.; Hoal, E.G.; Schurr, E. Neutrophils: Innate Effectors of TB Resistance? Front. Immunol. 2018, 9, 2637. [Google Scholar] [CrossRef] [Green Version]
  15. Silva-Sanchez, A.; Meza-Pérez, S.; Muñoz-Teneria, F.; Mata, D.; Calderon-Amador, J.; Estrada-Parra, S.; Hernández-Pando, R.; Estrada-García, I.; Flores-Romo, L. Mycobacterial Strains of Different Virulence Trigger Dissimilar Patterns of Immune System Activation In Vivo. In Understanding Tuberculosis-Deciphering the Secret Life of the Bacilli; InTech Open: London, UK, 2012. [Google Scholar]
  16. Scordo, J.M.; Knoell, D.L.; Torrelles, J.B. Alveolar epithelial cells in Mycobacterium tuberculosis infection: Active Players or Innocent Bystanders. J. Innate Immun. 2016, 8, 3–14. [Google Scholar] [CrossRef]
  17. Li, Y.; Wang, Y.; Liu, X. The role of airway epithelial cells in response to mycobacteria infection. Clin. Dev. Immunol. 2012, 2012, 791392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Faridgohar, M.; Nikoueinejad, H. New findings of Toll-like receptors involved in Mycobacterium tuberculosis infection. Pathog. Glob. Health 2017, 111, 256–264. [Google Scholar] [CrossRef] [PubMed]
  19. Diamond, C.E.; Khameneh, H.J.; Brough, D.; Mortellaro, A. Novel perspectives on non-canonical inflammasome activation. ImmunoTargets Ther. 2015, 4, 131. [Google Scholar]
  20. Dorhoi, A.; Nouailles, G.; Jörg, S.; Hagens, K.; Heinemann, E.; Pradl, L.; Oberbeck-Müller, D.; Duque-Correa, M.A.; Reece, S.T.; Ruland, J. Activation of the NLRP3 inflammasome by Mycobacterium tuberculosis is uncoupled from susceptibility to active tuberculosis. Eur. J. Immunol. 2012, 42, 374–384. [Google Scholar] [CrossRef] [PubMed]
  21. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Jo, E.-K.; Kim, J.K.; Shin, D.-M.; Sasakawa, C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell. Mol. Immunol. 2016, 13, 148–159. [Google Scholar] [CrossRef] [Green Version]
  23. Jayaraman, P.; Sada-Ovalle, I.; Nishimura, T.; Anderson, A.C.; Kuchroo, V.K.; Remold, H.G.; Behar, S.M. IL-1β promotes antimicrobial immunity in macrophages by regulating TNFR signaling and caspase-3 activation. J. Immunol. 2013, 190, 4196–4204. [Google Scholar] [CrossRef]
  24. Krishnan, N.; Robertson, B.D.; Thwaites, G. Pathways of IL-1β secretion by macrophages infected with clinical Mycobacterium tuberculosis strains. Tuberculosis 2013, 93, 538–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mishra, B.B.; Rathinam, V.A.K.; Martens, G.W.; Martinot, A.J.; Kornfeld, H.; Fitzgerald, K.A.; Sassetti, C.M. Nitric oxide controls the immunopathology of tuberculosis by inhibiting NLRP3 inflammasome-dependent processing of IL-1β. Nat. Immunol. 2013, 14, 52–60. [Google Scholar] [CrossRef] [Green Version]
  26. Ma, J.; Zhao, S.; Gao, X.; Wang, R.; Liu, J.; Zhou, X.; Zhou, Y. The roles of inflammasomes in host defense against Mycobacterium tuberculosis. Pathogens 2021, 10, 120. [Google Scholar] [CrossRef]
  27. Anochie, P.I.; Ndingkokhar, B.; Bueno, J.; Anyiam, F.E.; Ossai-Chidi, L.N.; Onyeneke, E.C.; Onyeozirila, A.C. African Medicinal Plants that Can Control or Cure Tuberculosis. Int. J. Pharm. Sci. Dev. Res. 2018, 4, 1–8. [Google Scholar]
  28. Rahman, F.; Hossan, S.; Mollik, A.; Islam, T.; Jahan, R.; Taufiq-Ur-Rahman, M.; Rahmatullah, M. Medicinal plants used against tuberculosis by traditional medicinal practitioners of Bogra district, Bangladesh. Planta Med. 2009, 75, PD64. [Google Scholar] [CrossRef]
  29. Gupta, V.K.; Kumar, M.M.; Bisht, D.; Kaushik, A. Plants in our combating strategies against Mycobacterium tuberculosis: Progress made and obstacles met. Pharm. Biol. 2017, 55, 1536–1544. [Google Scholar] [CrossRef] [Green Version]
  30. Semenya, S.S.; Maroyi, A. Medicinal plants used for the treatment of tuberculosis by Bapedi traditional healers in three districts of the Limpopo Province, South Africa. Afr. J. Tradit. Complement. Altern. Med. 2012, 10, 316–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Zhang, Q.; Jiang, X.; He, W.; Wei, K.; Sun, J.; Qin, X.; Zheng, Y.; Jiang, X. MCL Plays an Anti-Inflammatory Role in Mycobacterium tuberculosis Induced Immune Response by Inhibiting NF-κB and NLRP3 Inflammasome Activation. Mediat. Inflamm. 2017, 2017, 2432904. [Google Scholar] [CrossRef] [Green Version]
  32. Zhang, Q.; Sun, J.; Fu, Y.; He, W.; Li, Y.; Tan, H.; Xu, H.; Jiang, X. Guttiferone K Exerts the Anti-inflammatory Effect on Mycobacterium tuberculosis-(H37Ra-) Infected Macrophages by Targeting the TLR/IRAK-1 Mediated Akt and NF-κB Pathway. Mediat. Inflamm. 2020, 2020. [Google Scholar] [CrossRef]
  33. Madikizela, B.; Ndhlala, A.; Finnie, J.; Van Staden, J. Antimycobacterial, anti-inflammatory and genotoxicity evaluation of plants used for the treatment of tuberculosis and related symptoms in South Africa. J. Ethnopharmacol. 2014, 153, 386–391. [Google Scholar] [CrossRef]
  34. Zhang, Q.; Sun, J.; Wang, Y.; He, W.; Wang, L.; Zheng, Y.; Wu, J.; Zhang, Y.; Jiang, X. Antimycobacterial and Anti-inflammatory Mechanisms of Baicalin via Induced Autophagy in Macrophages Infected with Mycobacterium tuberculosis. Front. Microbiol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  35. He, W.; Sun, J.; Zhang, Q.; Li, Y.; Fu, Y.; Zheng, Y.; Jiang, X. Andrographolide exerts anti-inflammatory effects in Mycobacterium tuberculosis-infected macrophages by regulating the Notch1/Akt/NF-κB axis. J. Leukoc. Biol. 2020, 108, 1747–1764. [Google Scholar] [CrossRef]
  36. Velasco-Velázquez, M.A.; Barrera, D.; González-Arenas, A.; Rosales, C.; Agramonte-Hevia, J. Macrophage—Mycobacterium tuberculosis interactions: Role of complement receptor 3. Microb. Pathog. 2003, 35, 125–131. [Google Scholar] [CrossRef]
  37. Cywes, C.; Godenir, N.L.; Hoppe, H.C.; Scholle, R.R.; Steyn, L.M.; Kirsch, R.E.; Ehlers, M. Nonopsonic binding of Mycobacterium tuberculosis to human complement receptor type 3 expressed in Chinese hamster ovary cells. Infect. Immun. 1996, 64, 5373–5383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Maglione, P.J.; Xu, J.; Casadevall, A.; Chan, J. Fc gamma receptors regulate immune activation and susceptibility during Mycobacterium tuberculosis infection. J. Immunol. 2008, 180, 3329–3338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Melo, M.D.; Catchpole, I.R.; Haggar, G.; Stokes, R.W. Utilization of CD11b knockout mice to characterize the role of complement receptor 3 (CR3, CD11b/CD18) in the growth of Mycobacterium tuberculosis in macrophages. Cell. Immunol. 2000, 205, 13–23. [Google Scholar] [CrossRef]
  40. Schlesinger, L.S.; Azad, A.K.; Torrelles, J.B.; Roberts, E.; Vergne, I.; Deretic, V. Determinants of phagocytosis, phagosome biogenesis and autophagy for Mycobacterium tuberculosis. Handb. Tuberc. Immunol. Cell Biol. 2008, 1, 22. [Google Scholar]
  41. Schlesinger, L.S.; Bellinger-Kawahara, C.G.; Payne, N.R.; Horwitz, M.A. Phagocytosis of Mycobacterium tuberculosis is mediated by human monocyte complement receptors and complement component C3. J. Immunol. 1990, 144, 2771–2780. [Google Scholar]
  42. Ferguson, J.S.; Weis, J.J.; Martin, J.L.; Schlesinger, L.S. Complement protein C3 binding to Mycobacterium tuberculosis is initiated by the classical pathway in human bronchoalveolar lavage fluid. Infect. Immun. 2004, 72, 2564. [Google Scholar] [CrossRef] [Green Version]
  43. Armstrong, J.A.; Hart, P.D. Phagosome-lysosome interactions in cultured macrophages infected with virulent tubercle bacilli. Reversal of the usual nonfusion pattern and observations on bacterial survival. J. Exp. Med. 1975, 142, 1–16. [Google Scholar] [CrossRef] [Green Version]
  44. Kang, P.B.; Azad, A.K.; Torrelles, J.B.; Kaufman, T.M.; Beharka, A.; Tibesar, E.; DesJardin, L.E.; Schlesinger, L.S. The human macrophage mannose receptor directs Mycobacterium tuberculosis lipoarabinomannan-mediated phagosome biogenesis. J. Exp. Med. 2005, 202, 987–999. [Google Scholar] [CrossRef]
  45. Rajaram, M.V.S.; Arnett, E.; Azad, A.K.; Guirado, E.; Ni, B.; Gerberick, A.D.; He, L.Z.; Keler, T.; Thomas, L.J.; Lafuse, W.P.; et al. M. tuberculosis—Initiated Human Mannose Receptor Signaling Regulates Macrophage Recognition and Vesicle Trafficking by FcRγ-Chain, Grb2, and SHP-1. Cell Rep. 2017, 21, 126–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Garcia-Aguilar, T.; Espinosa-Cueto, P.; Magallanes-Puebla, A.; Mancilla, R. The Mannose Receptor Is Involved in the Phagocytosis of Mycobacteria-Induced Apoptotic Cells. J. Immunol. Res. 2016, 2016, 3845247. [Google Scholar] [CrossRef] [Green Version]
  47. Schlesinger, L.S.; Kaufman, T.M.; Iyer, S.; Hull, S.R.; Marchiando, L.K. Differences in mannose receptor-mediated uptake of lipoarabinomannan from virulent and attenuated strains of Mycobacterium tuberculosis by human macrophages. J. Immunol. 1996, 157, 4568–4575. [Google Scholar]
  48. Gaynor, C.D.; McCormack, F.X.; Voelker, D.R.; McGowan, S.E.; Schlesinger, L.S. Pulmonary surfactant protein A mediates enhanced phagocytosis of Mycobacterium tuberculosis by a direct interaction with human macrophages. J. Immunol. 1995, 155, 5343–5351. [Google Scholar] [PubMed]
  49. Pasula, R.; Downing, J.F.; Wright, J.R.; Kachel, D.L.; Davis, T.E., Jr.; Martin, W.J. Surfactant protein A (SP-A) mediates attachment of Mycobacterium tuberculosis to murine alveolar macrophages. Am. J. Respir. Cell Mol. Biol. 1997, 17, 209–217. [Google Scholar] [CrossRef] [PubMed]
  50. Van de Veerdonk, F.L.; Teirlinck, A.C.; Kleinnijenhuis, J.; Kullberg, B.J.; van Crevel, R.; van der Meer, J.W.; Joosten, L.A.; Netea, M.G. Mycobacterium tuberculosis induces IL-17A responses through TLR4 and dectin-1 and is critically dependent on endogenous IL-1. J. Leukoc. Biol. 2010, 88, 227–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Yadav, M.; Schorey, J.S. The β-glucan receptor dectin-1 functions together with TLR2 to mediate macrophage activation by mycobacteria. Blood 2006, 108, 3168–3175. [Google Scholar] [CrossRef]
  52. Kleinnijenhuis, J.; Joosten, L.A.; van de Veerdonk, F.L.; Savage, N.; van Crevel, R.; Kullberg, B.J.; van der Ven, A.; Ottenhoff, T.H.; Dinarello, C.A.; van der Meer, J.W. Transcriptional and inflammasome-mediated pathways for the induction of IL-1β production by Mycobacterium tuberculosis. Eur. J. Immunol. 2009, 39, 1914–1922. [Google Scholar] [CrossRef]
  53. Shams, H.; Wizel, B.; Lakey, D.L.; Samten, B.; Vankayalapati, R.; Valdivia, R.H.; Kitchens, R.L.; Griffith, D.E.; Barnes, P.F. The CD14 receptor does not mediate entry of Mycobacterium tuberculosis into human mononuclear phagocytes. FEMS Immunol. Med. Microbiol. 2003, 36, 63–69. [Google Scholar] [CrossRef] [Green Version]
  54. Yoshida, A.; Inagawa, H.; Kohchi, C.; Nishizawa, T.; Soma, G.-I. The role of toll-like receptor 2 in survival strategies of Mycobacterium tuberculosis in macrophage phagosomes. Anticancer Res. 2009, 29, 907–910. [Google Scholar] [PubMed]
  55. Sánchez, D.; Rojas, M.; Hernández, I.; Radzioch, D.; García, L.F.; Barrera, L.F. Role of TLR2-and TLR4-mediated signaling in Mycobacterium tuberculosis-induced macrophage death. Cell. Immunol. 2010, 260, 128–136. [Google Scholar] [CrossRef]
  56. Bermudez, L.E.; Goodman, J. Mycobacterium tuberculosis invades and replicates within type II alveolar cells. Infect. Immun. 1996, 64, 1400–1406. [Google Scholar] [CrossRef] [Green Version]
  57. Lee, H.M.; Yuk, J.M.; Shin, D.M.; Jo, E.K. Dectin-1 is inducible and plays an essential role for mycobacteria-induced innate immune responses in airway epithelial cells. J. Clin. Immunol. 2009, 29, 795–805. [Google Scholar] [CrossRef]
  58. Hadifar, S.; Behrouzi, A.; Fateh, A.; Khatami, S.; Jamnani, F.R.; Siadat, S.D.; Vaziri, F. Interruption of signaling pathways in lung epithelial cell by Mycobacterium tuberculosis. bioRxiv 2018. [Google Scholar] [CrossRef]
  59. Sequeira, P.C.; Senaratne, R.H.; Riley, L.W. Inhibition of toll-like receptor 2 (TLR-2)-mediated response in human alveolar epithelial cells by mycolic acids and Mycobacterium tuberculosis mce1 operon mutant. Pathog. Dis. 2014, 70, 132–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Mvubu, N.E.; Pillay, B.; McKinnon, L.R.; Pillay, M. Mycobacterium tuberculosis strains induce strain-specific cytokine and chemokine response in pulmonary epithelial cells. Cytokine 2018, 104, 53–64. [Google Scholar] [CrossRef] [PubMed]
  61. Troutman, T.D.; Bazan, J.F.; Pasare, C. Toll-like receptors, signaling adapters and regulation of the pro-inflammatory response by PI3K. Cell Cycle 2012, 11, 3559–3567. [Google Scholar] [CrossRef] [Green Version]
  62. Mvubu, N.E.; Pillay, B.; Gamieldien, J.; Bishai, W.; Pillay, M. Canonical pathways, networks and transcriptional factor regulation by clinical strains of Mycobacterium tuberculosis in pulmonary alveolar epithelial cells. Tuberculosis 2016, 97, 73–85. [Google Scholar] [CrossRef]
  63. Kim, K.; Sohn, H.; Kim, J.-S.; Choi, H.-G.; Byun, E.-H.; Lee, K.-I.; Shin, S.J.; Song, C.-H.; Park, J.-K.; Kim, H.-J. Mycobacterium tuberculosis Rv0652 stimulates production of tumour necrosis factor and monocytes chemoattractant protein-1 in macrophages through the Toll-like receptor 4 pathway. Immunology 2012, 136, 231–240. [Google Scholar] [CrossRef]
  64. Fremond, C.M.; Togbe, D.; Doz, E.; Rose, S.; Vasseur, V.; Maillet, I.; Jacobs, M.; Ryffel, B.; Quesniaux, V.F.J. IL-1 Receptor-Mediated Signal Is an Essential Component of MyD88-Dependent Innate Response to Mycobacterium tuberculosis Infection. J. Immunol. 2007, 179, 1178–1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Scanga, C.A.; Bafica, A.; Feng, C.G.; Cheever, A.W.; Hieny, S.; Sher, A. MyD88-deficient mice display a profound loss in resistance to Mycobacterium tuberculosis associated with partially impaired Th1 cytokine and nitric oxide synthase 2 expression. Infect. Immun. 2004, 72, 2400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sugawara, I.; Yamada, H.; Mizuno, S.; Takeda, K.; Akira, S. Mycobacterial infection in MyD88-deficient mice. Microbiol. Immunol. 2003, 47, 841–847. [Google Scholar] [CrossRef] [Green Version]
  67. Pellegrini, C.; Antonioli, L.; Lopez-Castejon, G.; Blandizzi, C.; Fornai, M. Canonical and non-canonical activation of NLRP3 inflammasome at the crossroad between immune tolerance and intestinal inflammation. Front. Immunol. 2017, 8, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Wawrocki, S.; Druszczynska, M. Inflammasomes in Mycobacterium tuberculosis-Driven Immunity. Can. J. Infect. Dis. Med. Microbiol. 2017, 2017, 2309478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Hosseinian, N.; Cho, Y.; Lockey, R.F.; Kolliputi, N. The role of the NLRP3 inflammasome in pulmonary diseases. Ther. Adv. Respir. Dis. 2015, 9, 188–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Lamkanfi, M.; Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157, 1013–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Sharma, D.; Kanneganti, T.-D. The cell biology of inflammasomes: Mechanisms of inflammasome activation and regulation. J. Cell Biol. 2016, 213, 617–629. [Google Scholar] [CrossRef] [Green Version]
  72. 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’Império-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. [Google Scholar] [CrossRef]
  73. Carmona, J.; Cruz, A.; Moreira-Teixeira, L.; Sousa, C.; Sousa, J.; Osorio, N.S.; Saraiva, A.L.; Svenson, S.; Kallenius, G.; Pedrosa, J. Mycobacterium tuberculosis strains are differentially recognized by TLRs with an impact on the immune response. PLoS ONE 2013, 8, e67277. [Google Scholar] [CrossRef] [Green Version]
  74. Amaral, E.P.; Andrade, B.d.B. Nuclear Factor κB Activation Pathways during Mycobacterium tuberculosis Infection. Clin. Med. Insights Case Rep. 2017, 9, 1–10. [Google Scholar]
  75. Pechkovsky, D.V.; Goldmann, T.; Vollmer, E.; Müller-Quernheim, J.; Zissel, G. Interleukin-18 expression by alveolar epithelial cells type II in tuberculosis and sarcoidosis. FEMS Immunol. Med. Microbiol. 2006, 46, 30–38. [Google Scholar] [CrossRef] [Green Version]
  76. Ramsugit, S.; Pillay, B.; Pillay, M. Evaluation of the role of Mycobacterium tuberculosis pili (MTP) as an adhesin, invasin, and cytokine inducer of epithelial cells. Braz. J. Infect. Dis. 2016, 20, 160–165. [Google Scholar] [CrossRef] [Green Version]
  77. Juffermans, N.P.; Florquin, S.; Camoglio, L.; Verbon, A.; Kolk, A.H.; Speelman, P.; van Deventer, S.J.H.; van der Poll, T. Interleukin-1 Signaling Is Essential for Host Defense during Murine Pulmonary Tuberculosis. J. Infect. Dis. 2000, 182, 902–908. [Google Scholar] [CrossRef] [PubMed]
  78. Jung, B.-G.; Vankayalapati, R.; Samten, B. Mycobacterium tuberculosis stimulates IL-1β production by macrophages in an ESAT-6 dependent manner with the involvement of serum amyloid A3. Mol. Immunol. 2021, 135, 285–293. [Google Scholar] [CrossRef] [PubMed]
  79. Yamada, H.; Mizumo, S.; Horai, R.; Iwakura, Y.; Sugawara, I. Protective role of interleukin-1 in mycobacterial infection in IL-1 α/β double-knockout mice. Lab. Investig. 2000, 80, 759–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Oh, C.; Verma, A.; Aachoui, Y. Caspase-11 Non-canonical Inflammasomes in the Lung. Front. Immunol. 2020, 11. [Google Scholar] [CrossRef]
  81. Chaves, M.M.; Sinflorio, D.A.; Thorstenberg, M.L.; Martins, M.D.A.; Moreira-Souza, A.C.A.; Rangel, T.P.; Silva, C.L.; Bellio, M.; Canetti, C.; Coutinho-Silva, R. Non-canonical NLRP3 inflammasome activation and IL-1β signaling are necessary to L. amazonensis control mediated by P2X7 receptor and leukotriene B4. PLoS Pathog. 2019, 15, e1007887. [Google Scholar] [CrossRef]
  82. Gutierrez, M.G.; Master, S.S.; Singh, S.B.; Taylor, G.A.; Colombo, M.I.; Deretic, V. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 2004, 119, 753–766. [Google Scholar] [CrossRef] [Green Version]
  83. Mishra, B.B.; Moura-Alves, P.; Sonawane, A.; Hacohen, N.; Griffiths, G.; Moita, L.F.; Anes, E. Mycobacterium tuberculosis protein ESAT-6 is a potent activator of the NLRP3/ASC inflammasome. Cell. Microbiol. 2010, 12, 1046–1063. [Google Scholar] [CrossRef]
  84. Schneider, B.E.; Korbel, D.; Hagens, K.; Koch, M.; Raupach, B.; Enders, J.; Kaufmann, S.H.; Mittrücker, H.W.; Schaible, U.E. A role for IL-18 in protective immunity against Mycobacterium tuberculosis. Eur. J. Immunol. 2010, 40, 396–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Nakanishi, K. Unique action of interleukin-18 on T cells and other immune cells. Front. Immunol. 2018, 9, 763. [Google Scholar] [CrossRef] [Green Version]
  86. Netea, M.G.; Kullberg, B.J.; Verschueren, I.; Meer, J.W.V.d. Interleukin-18 induces production of proinflammatory cytokines in mice: No intermediate role for the cytokines of the tumor necrosis factor family and interleukin-1β. Eur. J. Immunol. 2000, 30, 3057–3060. [Google Scholar] [CrossRef]
  87. Puren, A.J.; Fantuzzi, G.; Gu, Y.; Su, M.; Dinarello, C.A. Interleukin-18 (IFNgamma-inducing factor) induces IL-8 and IL-1beta via TNFalpha production from non-CD14+ human blood mononuclear cells. J. Clin. Investig. 1998, 101, 711–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Sugawara, I.; Yamada, H.; Kaneko, H.; Mizuno, S.; Takeda, K.; Akira, S. Role of interleukin-18 (IL-18) in mycobacterial infection in IL-18-gene-disrupted mice. Infect. Immun. 1999, 67, 2585–2589. [Google Scholar] [CrossRef] [Green Version]
  89. Kumar, N.P.; Moideen, K.; Banurekha, V.V.; Nair, D.; Babu, S. Plasma Proinflammatory Cytokines Are Markers of Disease Severity and Bacterial Burden in Pulmonary Tuberculosis. Open Forum Infect. Dis. 2019, 6, ofz257. [Google Scholar] [CrossRef]
  90. Chao, W.-C.; Yen, C.-L.; Hsieh, C.-Y.; Huang, Y.-F.; Tseng, Y.-L.; Nigrovic, P.A.; Shieh, C.-C. Mycobacterial infection induces higher interleukin-1β and dysregulated lung inflammation in mice with defective leukocyte NADPH oxidase. PLoS ONE 2017, 12, e0189453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Wang, Y.; Hu, C.; Wang, Z.; Kong, H.; Xie, W.; Wang, H. Serum IL-1β and IL-18 correlate with ESR and CRP in multidrug-resistant tuberculosis patients. J. Biomed. Res. 2015, 29, 426–428. [Google Scholar] [CrossRef] [Green Version]
  92. Santucci, N.; D’Attilio, L.; Besedovsky, H.; Del Rey, A.; Bay, M.L.; Bottasso, O. A clinical correlate of the dysregulated immunoendocrine response in human tuberculosis. Neuroimmunomodulation 2010, 17, 184–187. [Google Scholar] [CrossRef] [PubMed]
  93. Boldrin, F.; Provvedi, R.; Cioetto Mazzabò, L.; Segafreddo, G.; Manganelli, R. Tolerance and Persistence to Drugs: A Main Challenge in the Fight Against Mycobacterium tuberculosis. Front. Microbiol. 2020, 11, 1924. [Google Scholar] [CrossRef]
  94. Mandal, S.; Njikan, S.; Kumar, A.; Early, J.V.; Parish, T. The relevance of persisters in tuberculosis drug discovery. Microbiology 2019, 165, 492–499. [Google Scholar] [CrossRef]
  95. Sarathy, J.P.; Via, L.E.; Weiner, D.; Blanc, L.; Boshoff, H.; Eugenin, E.A.; Barry III, C.E.; Dartois, V.A. Extreme drug tolerance of Mycobacterium tuberculosis in caseum. Antimicrob. Agents Chemother. 2018, 62, e02266-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kahaliw, W.; Aseffa, A.; Abebe, M.; Teferi, M.; Engidawork, E. Evaluation of the antimycobacterial activity of crude extracts and solvent fractions of selected Ethiopian medicinal plants. BMC Complement. Altern. Med. 2017, 17, 143. [Google Scholar] [CrossRef] [Green Version]
  97. Gemechu, A.; Giday, M.; Worku, A.; Ameni, G. In vitro Anti-mycobacterial activity of selected medicinal plants against Mycobacterium tuberculosis and Mycobacterium bovis Strains. BMC Complement. Altern. Med. 2013, 13, 291. [Google Scholar] [CrossRef] [Green Version]
  98. Assam, J.P.A.; Tcham, M.F.Y.; Moni, N.E.D.F.; Betote, D.P.H.; Fossi, T.C.; Penlap, B.V. Phytochemical screening, Antimycobacterial activity of three medicinal Cameroonians plants and Acute toxicity of hydroethanolic extract of Vitellaria paradoxa. J. Drug Deliv. Ther. 2020, 10, 96–104. [Google Scholar] [CrossRef]
  99. Fyhrquist, P.; Laakso, I.; Marco, S.G.; Julkunen-Tiitto, R.; Hiltunen, R. Antimycobacterial activity of ellagitannin and ellagic acid derivate rich crude extracts and fractions of five selected species of Terminalia used for treatment of infectious diseases in African traditional medicine. S. Afr. J. Bot. 2014, 90, 1–16. [Google Scholar] [CrossRef] [Green Version]
  100. Putri, D.U.; Rintiswati, N.; Soesatyo, M.H.; Haryana, S.M. Immune modulation properties of herbal plant leaves: Phyllanthus niruri aqueous extract on immune cells of tuberculosis patient—In vitro study. Nat. Prod. Res. 2018, 32, 463–467. [Google Scholar] [CrossRef] [PubMed]
  101. Krug, S.; Parveen, S.; Bishai, W.R. Host-Directed Therapies: Modulating Inflammation to Treat Tuberculosis. Front. Immunol. 2021, 12. [Google Scholar] [CrossRef]
  102. Zumla, A.; Rao, M.; Dodoo, E.; Maeurer, M. Potential of immunomodulatory agents as adjunct host-directed therapies for multidrug-resistant tuberculosis. BMC Med. 2016, 14, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ahmed, S.; Raqib, R.; Guðmundsson, G.H.; Bergman, P.; Agerberth, B.; Rekha, R.S. Host-Directed Therapy as a Novel Treatment Strategy to Overcome Tuberculosis: Targeting Immune Modulation. Antibiotics 2020, 9, 21. [Google Scholar] [CrossRef] [Green Version]
  104. Ji, Q.; Ding, Y.-H.; Sun, Y.; Zhang, Y.; Gao, H.-E.; Song, H.-N.; Yang, M.; Liu, X.-L.; Zhang, Z.-X.; Li, Y.-H.; et al. Antineoplastic effects and mechanisms of micheliolide in acute myelogenous leukemia stem cells. Oncotarget 2016, 7, 65012–65023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Viennois, E.; Xiao, B.; Ayyadurai, S.; Wang, L.; Wang, P.G.; Zhang, Q.; Chen, Y.; Merlin, D. Micheliolide, a new sesquiterpene lactone that inhibits intestinal inflammation and colitis-associated cancer. Lab. Investig. 2014, 94, 950–965. [Google Scholar] [CrossRef] [Green Version]
  106. Ivanescu, B.; Miron, A.; Corciova, A. Sesquiterpene lactones from Artemisia genus: Biological activities and methods of analysis. J. Anal. Methods Chem. 2015, 2015. [Google Scholar] [CrossRef] [Green Version]
  107. Picman, A.K. Biological activities of sesquiterpene lactones. Biochem. Syst. Ecol. 1986, 14, 255–281. [Google Scholar] [CrossRef]
  108. Qin, X.; Jiang, X.; Jiang, X.; Wang, Y.; Miao, Z.; He, W.; Yang, G.; Lv, Z.; Yu, Y.; Zheng, Y. Micheliolide inhibits LPS-induced inflammatory response and protects mice from LPS challenge. Sci. Rep. 2016, 6, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Xu, G.; Feng, C.; Zhou, Y.; Han, Q.-B.; Qiao, C.-F.; Huang, S.-X.; Chang, D.C.; Zhao, Q.-S.; Luo, K.Q.; Xu, H.-X. Bioassay and ultraperformance liquid chromatography/mass spectrometry guided isolation of apoptosis-inducing benzophenones and xanthone from the pericarp of Garcinia yunnanensis Hu. J. Agric. Food Chem. 2008, 56, 11144–11150. [Google Scholar] [CrossRef]
  110. Li, H.; Meng, X.-x.; Zhang, L.; Zhang, B.-j.; Liu, X.-y.; Fu, W.-w.; Tan, H.-s.; Lao, Y.-z.; Xu, H.-x. Oblongifolin C and guttiferone K extracted from Garcinia yunnanensis fruit synergistically induce apoptosis in human colorectal cancer cells in vitro. Acta Pharmacol. Sin. 2017, 38, 252–263. [Google Scholar] [CrossRef]
  111. Pu, Z.; Han, C.; Zhang, W.; Xu, M.; Wu, Z.; Liu, Y.; Wu, M.; Sun, H.; Xie, H. Systematic understanding of the mechanism and effects of Arctigenin attenuates inflammation in dextran sulfate sodium-induced acute colitis through suppression of NLRP3 inflammasome by SIRT1. Am. J. Transl. Res. 2019, 11, 3992. [Google Scholar]
  112. Yuan, R.; Fan, H.; Cheng, S.; Gao, W.; Xu, X.; Lv, S.; Ye, M.; Wu, M.; Zhu, X.; Zhang, Y. Silymarin prevents NLRP3 inflammasome activation and protects against intracerebral hemorrhage. Biomed. Pharmacother. 2017, 93, 308–315. [Google Scholar] [CrossRef]
  113. Hu, Q.-H.; Zhang, X.; Pan, Y.; Li, Y.-C.; Kong, L.-D. Allopurinol, quercetin and rutin ameliorate renal NLRP3 inflammasome activation and lipid accumulation in fructose-fed rats. Biochem. Pharmacol. 2012, 84, 113–125. [Google Scholar] [CrossRef]
  114. Wang, S.; Wang, J.; Wei, H.; Gu, T.; Wang, J.; Wu, Z.; Yang, Q. Genistein attenuates acute cerebral ischemic damage by inhibiting the NLRP3 inflammasome in reproductively senescent mice. Front. Aging Neurosci. 2020, 12, 153. [Google Scholar] [CrossRef] [PubMed]
  115. Budai, M.M.; Varga, A.; Milesz, S.; Tőzsér, J.; Benkő, S. Aloe vera downregulates LPS-induced inflammatory cytokine production and expression of NLRP3 inflammasome in human macrophages. Mol. Immunol. 2013, 56, 471–479. [Google Scholar] [CrossRef] [Green Version]
  116. Gong, Q.; He, L.-L.; Wang, M.-L.; Ouyang, H.; Gao, H.-W.; Feng, Y.-L.; Yang, S.-L.; Du, L.-J.; Li, J.; Luo, Y.-Y. Anemoside B4 protects rat kidney from adenine-induced injury by attenuating inflammation and fibrosis and enhancing podocin and nephrin expression. Evid. Based Complement. Altern. Med. 2019, 2019. [Google Scholar] [CrossRef] [PubMed]
  117. Tsai, P.-Y.; Ka, S.-M.; Chang, J.-M.; Chen, H.-C.; Shui, H.-A.; Li, C.-Y.; Hua, K.-F.; Chang, W.-L.; Huang, J.-J.; Yang, S.-S. Epigallocatechin-3-gallate prevents lupus nephritis development in mice via enhancing the Nrf2 antioxidant pathway and inhibiting NLRP3 inflammasome activation. Free Radic. Biol. Med. 2011, 51, 744–754. [Google Scholar] [CrossRef] [PubMed]
  118. Ding, S.; Wang, H.; Wang, M.; Bai, L.; Yu, P.; Wu, W. Resveratrol alleviates chronic “real-world” ambient particulate matter-induced lung inflammation and fibrosis by inhibiting NLRP3 inflammasome activation in mice. Ecotoxicol. Environ. Saf. 2019, 182, 109425. [Google Scholar] [CrossRef] [PubMed]
  119. Matias, M.L.; Gomes, V.J.; Romao-Veiga, M.; Ribeiro, V.R.; Nunes, P.R.; Romagnoli, G.G.; Peracoli, J.C.; Peracoli, M.T.S. Silibinin downregulates the NF-κB pathway and NLRP1/NLRP3 inflammasomes in monocytes from pregnant women with preeclampsia. Molecules 2019, 24, 1548. [Google Scholar] [CrossRef] [Green Version]
  120. Wen, Y.; Pan, M.M.; Lv, L.L.; Tang, T.T.; Zhou, L.T.; Wang, B.; Liu, H.; Wang, F.M.; Ma, K.L.; Tang, R.N. Artemisinin attenuates tubulointerstitial inflammation and fibrosis via the NF-κB/NLRP3 pathway in rats with 5/6 subtotal nephrectomy. J. Cell. Biochem. 2019, 120, 4291–4300. [Google Scholar] [CrossRef]
  121. Zhang, L.; Wang, X.Z.; Li, Y.S.; Zhang, L.; Hao, L.R. Icariin ameliorates IgA nephropathy by inhibition of nuclear factor kappa b/Nlrp3 pathway. FEBS Open Bio 2017, 7, 54–63. [Google Scholar] [CrossRef] [Green Version]
  122. Tang, J.; Li, Y.; Wang, J.; Wu, Q.; Yan, H. Polydatin suppresses the development of lung inflammation and fibrosis by inhibiting activation of the NACHT domain-, leucine-rich repeat-, and pyd-containing protein 3 inflammasome and the nuclear factor-κB pathway after Mycoplasma pneumoniae infection. J. Cell. Biochem. 2019, 120, 10137–10144. [Google Scholar] [CrossRef]
  123. Kang, L.-L.; Zhang, D.-M.; Ma, C.-H.; Zhang, J.-H.; Jia, K.-K.; Liu, J.-H.; Wang, R.; Kong, L.-D. Cinnamaldehyde and allopurinol reduce fructose-induced cardiac inflammation and fibrosis by attenuating CD36-mediated TLR4/6-IRAK4/1 signaling to suppress NLRP3 inflammasome activation. Sci. Rep. 2016, 6, 1–18. [Google Scholar] [CrossRef]
  124. Han, W.; Ma, Q.; Liu, Y.; Wu, W.; Tu, Y.; Huang, L.; Long, Y.; Wang, W.; Yee, H.; Wan, Z.; et al. Huangkui capsule alleviates renal tubular epithelial-mesenchymal transition in diabetic nephropathy via inhibiting NLRP3 inflammasome activation and TLR4/NF-κB signaling. Phytomedicine 2019, 57, 203–214. [Google Scholar] [CrossRef]
  125. Chakravarti, R.N.; Chakravarti, D. Andrographolide, the active constituent of Andrographis paniculata Nees; a preliminary communication. Ind. Med. Gaz. 1951, 86, 96–97. [Google Scholar]
  126. Okhuarobo, A.; Falodun, J.E.; Erharuyi, O.; Imieje, V.; Falodun, A.; Langer, P. Harnessing the medicinal properties of Andrographis paniculata for diseases and beyond: A review of its phytochemistry and pharmacology. Asian Pac. J. Trop. Dis. 2014, 4, 213–222. [Google Scholar] [CrossRef]
  127. Li, C.-W.; Deng, M.-Z.; Gao, Z.-J.; Dang, Y.-Y.; Zheng, G.-D.; Yang, X.-J.; Chao, Y.-X.; Cai, Y.-F.; Wu, X.-L. Effects of compound K, a metabolite of ginsenosides, on memory and cognitive dysfunction in db/db mice involve the inhibition of ER stress and the NLRP3 inflammasome pathway. Food Funct. 2020, 11, 4416–4427. [Google Scholar] [CrossRef]
  128. Chen, W.; Wang, J.; Luo, Y.; Wang, T.; Li, X.; Li, A.; Li, J.; Liu, K.; Liu, B. Ginsenoside Rb1 and compound K improve insulin signaling and inhibit ER stress-associated NLRP3 inflammasome activation in adipose tissue. J. Ginseng Res. 2016, 40, 351–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Gong, Z.; Zhou, J.; Li, H.; Gao, Y.; Xu, C.; Zhao, S.; Chen, Y.; Cai, W.; Wu, J. Curcumin suppresses NLRP3 inflammasome activation and protects against LPS-induced septic shock. Mol. Nutr. Food Res. 2015, 59, 2132–2142. [Google Scholar] [CrossRef] [PubMed]
  130. Yu, S.-X.; Du, C.-T.; Chen, W.; Lei, Q.-Q.; Li, N.; Qi, S.; Zhang, X.-J.; Hu, G.-Q.; Deng, X.-M.; Han, W.-Y. Genipin inhibits NLRP3 and NLRC4 inflammasome activation via autophagy suppression. Sci. Rep. 2015, 5, 1–12. [Google Scholar] [CrossRef] [Green Version]
  131. Pan, C.-W.; Pan, Z.-Z.; Hu, J.-J.; Chen, W.-L.; Zhou, G.-Y.; Lin, W.; Jin, L.-X.; Xu, C.-L. Mangiferin alleviates lipopolysaccharide and D-galactosamine-induced acute liver injury by activating the Nrf2 pathway and inhibiting NLRP3 inflammasome activation. Eur. J. Pharmacol. 2016, 770, 85–91. [Google Scholar] [CrossRef]
  132. Qiu, H.; Liu, W.; Lan, T.; Pan, W.; Chen, X.; Wu, H.; Xu, D. Salvianolate reduces atrial fibrillation through suppressing atrial interstitial fibrosis by inhibiting TGF-β1/Smad2/3 and TXNIP/NLRP3 inflammasome signaling pathways in post-MI rats. Phytomedicine 2018, 51, 255–265. [Google Scholar] [CrossRef]
  133. Hua, K.F.; Chou, J.C.; Ka, S.M.; Tasi, Y.L.; Chen, A.; Wu, S.H.; Chiu, H.W.; Wong, W.T.; Wang, Y.F.; Tsai, C.L.; et al. Cyclooxygenase-2 regulates NLRP3 inflammasome-derived IL-1β production. J. Cell Physiol. 2015, 230, 863–874. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, G.; Jang, J.H.; Kim, S.W.; Han, S.-H.; Ma, K.-H.; Jang, J.-K.; Kang, H.C.; Cho, Y.-Y.; Lee, H.S.; Lee, J.Y. Sweroside prevents non-alcoholic steatohepatitis by suppressing activation of the NLRP3 inflammasome. Int. J. Mol. Sci. 2020, 21, 2790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. He, H.; Jiang, H.; Chen, Y.; Ye, J.; Wang, A.; Wang, C.; Liu, Q.; Liang, G.; Deng, X.; Jiang, W. Oridonin is a covalent NLRP3 inhibitor with strong anti-inflammasome activity. Nat. Commun. 2018, 9, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Gao, Y.; Lv, X.; Yang, H.; Peng, L.; Ci, X. Isoliquiritigenin exerts antioxidative and anti-inflammatory effects via activating the KEAP-1/Nrf2 pathway and inhibiting the NF-κB and NLRP3 pathways in carrageenan-induced pleurisy. Food Funct. 2020, 11, 2522–2534. [Google Scholar] [CrossRef]
  137. Wang, Z.; Xu, G.; Gao, Y.; Zhan, X.; Qin, N.; Fu, S.; Li, R.; Niu, M.; Wang, J.; Liu, Y. Cardamonin from a medicinal herb protects against LPS-induced septic shock by suppressing NLRP3 inflammasome. Acta Pharm. Sin. B 2019, 9, 734–744. [Google Scholar] [CrossRef] [PubMed]
  138. Han, X.; Song, J.; Lian, L.-H.; Yao, Y.-L.; Shao, D.-Y.; Fan, Y.; Hou, L.-S.; Wang, G.; Zheng, S.; Wu, Y.-L. Ginsenoside 25-och3-ppd promotes activity of lxrs to ameliorate p2x7r-mediated nlrp3 inflammasome in the development of hepatic fibrosis. J. Agric. Food Chem. 2018, 66, 7023–7035. [Google Scholar] [CrossRef] [PubMed]
  139. Jiang, J.; Sun, X.; Akther, M.; Lian, M.-L.; Quan, L.-H.; Koppula, S.; Han, J.-H.; Kopalli, S.R.; Kang, T.-B.; Lee, K.-H. Ginsenoside metabolite 20 (S)-protopanaxatriol from Panax ginseng attenuates inflammation-mediated NLRP3 inflammasome activation. J. Ethnopharmacol. 2020, 251, 112564. [Google Scholar] [CrossRef]
  140. Shen, X.; Dong, X.; Han, Y.; Li, Y.; Ding, S.; Zhang, H.; Sun, Z.; Yin, Y.; Li, W.; Li, W. Ginsenoside Rg1 ameliorates glomerular fibrosis during kidney aging by inhibiting NOX4 and NLRP3 inflammasome activation in SAMP8 mice. Int. Immunopharmacol. 2020, 82, 106339. [Google Scholar] [CrossRef]
  141. Shi, Y.; Wang, H.; Zheng, M.; Xu, W.; Yang, Y.; Shi, F. Ginsenoside Rg3 suppresses the NLRP3 inflammasome activation through inhibition of its assembly. FASEB J. 2020, 34, 208–221. [Google Scholar] [CrossRef] [Green Version]
  142. Li, R.; Lu, K.; Wang, Y.; Chen, M.; Zhang, F.; Shen, H.; Yao, D.; Gong, K.; Zhang, Z. Triptolide attenuates pressure overload-induced myocardial remodeling in mice via the inhibition of NLRP3 inflammasome expression. Biochem. Biophys. Res. Commun. 2017, 485, 69–75. [Google Scholar] [CrossRef]
  143. Yan, T.; Wang, H.; Cao, L.; Wang, Q.; Takahashi, S.; Yagai, T.; Li, G.; Krausz, K.W.; Wang, G.; Gonzalez, F.J. Glycyrrhizin alleviates nonalcoholic steatohepatitis via modulating bile acids and meta-inflammation. Drug Metab. Dispos. 2018, 46, 1310–1319. [Google Scholar] [CrossRef]
  144. Cui, L.; Li, C.; Zhuo, Y.; Yang, L.; Cui, N.; Li, Y.; Zhang, S. Saikosaponin A inhibits the activation of pancreatic stellate cells by suppressing autophagy and the NLRP3 inflammasome via the AMPK/mTOR pathway. Biomed. Pharmacother. 2020, 128, 110216. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, S.; Tian, L.; Chai, G.; Wen, B.; Wang, B. Targeting heme oxygenase-1 by quercetin ameliorates alcohol-induced acute liver injury via inhibiting NLRP3 inflammasome activation. Food Funct. 2018, 9, 4184–4193. [Google Scholar] [CrossRef] [PubMed]
  146. Ding, T.; Wang, S.; Zhang, X.; Zai, W.; Fan, J.; Chen, W.; Bian, Q.; Luan, J.; Shen, Y.; Zhang, Y. Kidney protection effects of dihydroquercetin on diabetic nephropathy through suppressing ROS and NLRP3 inflammasome. Phytomedicine 2018, 41, 45–53. [Google Scholar] [CrossRef] [PubMed]
  147. Li, X.; Mei, W.; Huang, Z.; Zhang, L.; Xu, B.; Shi, X.; Xiao, Y.; Ma, Z.; Liao, T.; Zhang, H. Casticin suppresses monoiodoacetic acid-induced knee osteoarthritis through inhibiting HIF-1α/NLRP3 inflammasome signaling. Int. Immunopharmacol. 2020, 86, 106745. [Google Scholar] [CrossRef]
  148. Cui, D.; Liu, S.; Tang, M.; Lu, Y.; Zhao, M.; Mao, R.; Wang, C.; Yuan, Y.; Li, L.; Chen, Y. Phloretin ameliorates hyperuricemia-induced chronic renal dysfunction through inhibiting NLRP3 inflammasome and uric acid reabsorption. Phytomedicine 2020, 66, 153111. [Google Scholar] [CrossRef]
  149. Wang, Y.-J.; Chen, Y.-Y.; Hsiao, C.-M.; Pan, M.-H.; Wang, B.-J.; Chen, Y.-C.; Ho, C.-T.; Huang, K.-C.; Chen, R.-J. Induction of autophagy by pterostilbene contributes to the prevention of renal fibrosis via attenuating NLRP3 inflammasome activation and epithelial-mesenchymal transition. Front. Cell Dev. Biol. 2020, 8, 436. [Google Scholar] [CrossRef]
  150. Xin, R.; Sun, X.; Wang, Z.; Yuan, W.; Jiang, W.; Wang, L.; Xiang, Y.; Zhang, H.; Li, X.; Hou, Y. Apocynin inhibited NLRP3/XIAP signalling to alleviate renal fibrotic injury in rat diabetic nephropathy. Biomed. Pharmacother. 2018, 106, 1325–1331. [Google Scholar] [CrossRef]
  151. Liang, Q.; Cai, W.; Zhao, Y.; Xu, H.; Tang, H.; Chen, D.; Qian, F.; Sun, L. Lycorine ameliorates bleomycin-induced pulmonary fibrosis via inhibiting NLRP3 inflammasome activation and pyroptosis. Pharmacol. Res. 2020, 158, 104884. [Google Scholar] [CrossRef]
  152. Mahzari, A.; Li, S.; Zhou, X.; Li, D.; Fouda, S.; Alhomrani, M.; Alzahrani, W.; Robinson, S.R.; Ye, J.-M. Matrine protects against MCD-induced development of NASH via upregulating HSP72 and downregulating mTOR in a manner distinctive from metformin. Front. Pharmacol. 2019, 10, 405. [Google Scholar] [CrossRef]
  153. Wu, X.; Zhang, F.; Xiong, X.; Lu, C.; Lian, N.; Lu, Y.; Zheng, S. Tetramethylpyrazine reduces inflammation in liver fibrosis and inhibits inflammatory cytokine expression in hepatic stellate cells by modulating NLRP 3 inflammasome pathway. IUBMB Life 2015, 67, 312–321. [Google Scholar] [CrossRef]
  154. Liu, G.; Shi, Y.; Peng, X.; Liu, H.; Peng, Y.; He, L. Astaxanthin attenuates adriamycin-induced focal segmental glomerulosclerosis. Pharmacology 2015, 95, 193–200. [Google Scholar] [CrossRef]
  155. Wang, L.; Ma, J.; Guo, C.; Chen, C.; Yin, Z.; Zhang, X.; Chen, X. Danggui buxue tang attenuates tubulointerstitial fibrosis via suppressing NLRP3 inflammasome in a rat model of unilateral ureteral obstruction. BioMed Res. Int. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
  156. Park, J.E.; Lee, H.; Rho, H.; Hong, S.M.; Kim, S.Y.; Lim, Y. Effect of Quamoclit angulata extract supplementation on oxidative stress and inflammation on hyperglycemia-induced renal damage in type 2 diabetic mice. Antioxidants 2020, 9, 459. [Google Scholar] [CrossRef]
  157. Yu, C.; He, Q.; Zheng, J.; Li, L.Y.; Hou, Y.H.; Song, F.Z. Sulforaphane improves outcomes and slows cerebral ischemic/reperfusion injury via inhibition of NLRP3 inflammasome activation in rats. Int. Immunopharmacol. 2017, 45, 74–78. [Google Scholar] [CrossRef] [PubMed]
  158. Kim, J.; Ahn, H.; Han, B.-C.; Shin, H.; Kim, J.-C.; Jung, E.-M.; Kim, J.; Yang, H.; Lee, J.; Kang, S.G. Obovatol inhibits NLRP3, AIM2, and non-canonical inflammasome activation. Phytomedicine 2019, 63, 153019. [Google Scholar] [CrossRef] [PubMed]
  159. Zhou, H.; Feng, L.; Xu, F.; Sun, Y.; Ma, Y.; Zhang, X.; Liu, H.; Xu, G.; Wu, X.; Shen, Y. Berberine inhibits palmitate-induced NLRP3 inflammasome activation by triggering autophagy in macrophages: A new mechanism linking berberine to insulin resistance improvement. Biomed. Pharmacother. 2017, 89, 864–874. [Google Scholar] [CrossRef]
  160. Bagherniya, M.; Khedmatgozar, H.; Fakheran, O.; Xu, S.; Johnston, T.P.; Sahebkar, A. Medicinal plants and bioactive natural products as inhibitors of NLRP3 inflammasome. Phytother. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  161. Liu, B.; Yu, J. Anti-NLRP3 Inflammasome Natural Compounds: An Update. Biomedicines 2021, 9, 136. [Google Scholar] [CrossRef]
  162. Tőzsér, J.; Benkő, S. Natural compounds as regulators of NLRP3 inflammasome-mediated IL-1β production. Mediat. Inflamm. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
  163. Ding, N.; Wei, B.; Fu, X.; Wang, C.; Wu, Y. Natural Products that Target the NLRP3 Inflammasome to Treat Fibrosis. Front. Pharmacol. 2020, 11, 2039. [Google Scholar] [CrossRef]
Figure 1. Overview of the alveolar NLRP3 inflammasome activation on alveolar innate cells during M. tuberculosis infection. The host PRRs (TLR4) is activated by M. tuberculosis surface PAMPs stimulate the NF-kB transcriptional factor to transcribe IL1, IL18, and CASP4/11 that are processed by activation of the inflammasome complex through NLRP3 and CASP1. Image was created in BioRender.com (accessed on 23 August 2021).
Figure 1. Overview of the alveolar NLRP3 inflammasome activation on alveolar innate cells during M. tuberculosis infection. The host PRRs (TLR4) is activated by M. tuberculosis surface PAMPs stimulate the NF-kB transcriptional factor to transcribe IL1, IL18, and CASP4/11 that are processed by activation of the inflammasome complex through NLRP3 and CASP1. Image was created in BioRender.com (accessed on 23 August 2021).
Ijms 22 09497 g001
Figure 2. Medicinal plant and/or bioactive regulators of the NLRP3 inflammasome that can be exploited as immunomodulatory host-directed adjuvants during M. tuberculosis infection. Compounds denoted with * were tested in vivo. Image was created in BioRender.com (accessed on 23 August 2021).
Figure 2. Medicinal plant and/or bioactive regulators of the NLRP3 inflammasome that can be exploited as immunomodulatory host-directed adjuvants during M. tuberculosis infection. Compounds denoted with * were tested in vivo. Image was created in BioRender.com (accessed on 23 August 2021).
Ijms 22 09497 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mvubu, N.E.; Chiliza, T.E. Exploring the Use of Medicinal Plants and Their Bioactive Derivatives as Alveolar NLRP3 Inflammasome Regulators during Mycobacterium tuberculosis Infection. Int. J. Mol. Sci. 2021, 22, 9497. https://doi.org/10.3390/ijms22179497

AMA Style

Mvubu NE, Chiliza TE. Exploring the Use of Medicinal Plants and Their Bioactive Derivatives as Alveolar NLRP3 Inflammasome Regulators during Mycobacterium tuberculosis Infection. International Journal of Molecular Sciences. 2021; 22(17):9497. https://doi.org/10.3390/ijms22179497

Chicago/Turabian Style

Mvubu, Nontobeko E., and Thamsanqa E. Chiliza. 2021. "Exploring the Use of Medicinal Plants and Their Bioactive Derivatives as Alveolar NLRP3 Inflammasome Regulators during Mycobacterium tuberculosis Infection" International Journal of Molecular Sciences 22, no. 17: 9497. https://doi.org/10.3390/ijms22179497

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop