Get Permission Ansari, Paul, D’Lima, Parackal, and Thomas: The Inflammasomes: A confounding chapter in Periodontics


Introduction

In the last two decades, substantial progress has been made regarding the understanding of mechanisms involved in the sensing of microbial pathogens by innate immune cells that form the first line of defense. Considerable research has been conducted in connection with the pathways that translate into the signal transduction and transcription of certain antimicrobial molecules and the means by which they activate adaptive immune responses. The innate immune system recognizes the infection and initiates responses for eliminating pathogens and repairing damaged cells. One significant multiprotein complex identified in these processes is the ‘inflammasome’. Despite the fact that inflammasome activation is an integral part of the innate immunity, overt inflammasome activation has the potential to give rise to many autoimmune and metabolic disorders. This necessitates the importance of understanding the processes involved here in physiological and pathological contexts to grasp the science behind inflammasome biology.

What is an Inflammasome?

Inflammasomes are the central signalling regulators of the innate immune system that rapidly recognize and trigger the body's response to infections and foreign substances that are potentially harmful to the host. They usually consist of a sensor protein (PRR), the adaptor protein apoptosis-associated speck-like protein (ASC) containing a caspase-recruitment domain (CARD), and the pro-inflammatory caspase -1.1 Following this, a vast array of physiological and pathogenic stimuli activates pro-caspase-1 that causes the cleavage of pro-inflammatory cytokines like IL-1β and IL-18.2 These cytokines which are extremely powerful molecules with a myriad of functions are regarded as an inevitable part of the host response in inflammation and the development of periodontal diseases.

Mechanism of action

Its mechanism of action is mediated by the PRR connecting to the ASC via oligomerization with its ligand followed by ASC converting pro-caspase-1 to its active form of caspase-1 with its adaptor CARD.

They are activated during inflammation by a vast array of microbial components, like evolutionarily conserved microbial structures, known as pathogen‐associated molecular patterns (PAMP) from bacteria, viruses or protozoans and the various endogenous molecules generated during tissue/cell damage such as ATP, uric acid crystals, and amyloid-b fibrils are called damage‐associated molecular patterns (DAMP). These maybe host derived or environmentally derived. The germline encoded pattern recognition receptors (PRR) which are found both in immune and non-immune cells then recognise these molecules. Some of these receptors assemble inflammasomes that activate cellular caspases which, thereby, induce the maturation of pro-inflammatory cytokines into their biologically active forms along with the induction of inflammation‐induced programmed cell death (pyroptosis). This, provides the host cell with dual defense mechanisms. Pyroptosis is a process occuring via an activating cleavage of cytoplasmic protein gasdermin D, which forms pores in the plasma membrane. Although it had been revealed in the past that caspase-1 activation resulted in cytokine maturation and pyroptosis, the details of the pathways involved were actually explored only later in a seminal paper by Martinon et al (2002) with the discovery of the inflammasome.

Now, a tight regulation to maintain a balance in inflammasome activation is of utmost importance. Inflammasome activation is usually a two-step process except in human monocytes (Figure 1):3

1) The first step is cell priming.

• This includes the recognition of various PAMPs or DAMPS that interact with a subset of many toll-like receptors, NOD1 or NOD2, or by the cytokines, tumor necrosis factor and IL-1β, resulting in transcriptional upregulation of inflammasome components by activating the transcription factor nuclear factor-κB.

• One goal here is to upregulate inflammasome components like the sensing PRR, caspase-1 and IL-1β through transcription and translation.

• The second goal here is the post-translational modification of the PRR and adaptor molecule ASC.

2) The second step is identifying a PAMP or DAMP specific to each inflammasome, which then induces the formation and activation of inflammasomes.

• Inflammasomes are named based on their respective PRRs that induce their activation. A number of PRR families, including a nucleotide-binding domain, leucine-rich repeat-containing proteins (NLR, also known as NOD-like receptors), absent in melanoma 2 (AIM2)-like receptors (ALRs), toll-like receptor system (TLRs), C-type lectin receptors (CLRs) and retinoic acid-inducible gene I (Rig-I)-like receptors (RLR) have been identified.

• The earliest family of PRRs to be identified were toll-like receptors (TLRs) which were observed either on the cell surface or in intracellular compartments. RLRs and NLRs unlike TLRs and CLRs, are cytosolic proteins that recognise intracellular microbial molecules or danger signals.

Figure 1

Inflammasome priming and activation. Inflammasomes should be primed before activation. Priming involves an increased expression of inflammasome components (IL1B, IL18, CAS1 AIM2, NLRP3, IFI16) induced by lipopolysaccharide or tumor necrosis factor-β via nuclear factor-κB-activation. After this, each inflammasome is activated resulting in the formation of unique inflammasome complexes. This is followed by oligomerization of PRRs and subsequent recruitment of ASC to activate caspase-1. This caspase-1 then converts proIL-1β and proIL-18 into their mature forms (IL-1β and IL-18) which is then released by cells via gasdermin D-pores.

https://s3-us-west-2.amazonaws.com/typeset-prod-media-server/3af09e0f-4506-4bc8-a0ad-6d8dc334b674image2.png

The NLR family

The pioneer family of sensor proteins that were found to assemble inflammasomes is the NOD-like receptor (or nucleotide binding domain and leucine-rich repeat containing receptor; NLR) family which is involved in the regulation of innate immune responses.4, 5 The NLR family was previously known as CATERPILLARs, NODs and NACHT leucine- rich repeats.4

Their members share a central nucleotide-binding domain (NBD) with most of them showing a C-terminal leucine-rich repeat (LRR) domain and a variable N-terminal domain. They are divided into four subfamilies depending upon their N-terminal domains. These four N-terminal domains include: (a) acidic transactivator domain (NLRA), (b) baculoviral inhibition of apoptosis protein repeat (BIR)-like domain (NLRB), (c) CARD and (d) pyrin domain. All NLRs except NLRP10 are categorized according to their domain structure and has a leucine-rich repeat (LRR) domain, a nucleotide binding domain (NBD), and a signalling domain.4 The signalling domain either directly, through a CARD or indirectly through a PYRIN domain causes the recruitment of caspase-1. Hence, they can be further grouped into NLRP or NLRC on the basis of whether the N terminus has a pyrin or caspase activation and recruitment domain (CARD), respectively. Certain members here like NLRP1, NLRP3, and NLRC4, have been understood as capable of forming inflammasomes while others, like NLRP6 and NLRP12, are still considered putative. Furthermore, newer families involved in forming inflammasomes such as the AIM2-like receptor (ALR) family,6 and the RIG-I-like receptor (RLR) family were discovered recently.7

NOD receptors not only sense microbes and damaged cells but also promote activation of NLRP3 and NLRP1 inflammasomes. The association between NOD2 mutations and the increased susceptibility to the development of Crohn's disease, an autoinflammatory disorder of the gastrointestinal tract signified the importance of NOD2 in inflammatory disease processes. In addition, recently an association between inflammatory bowel disease and an increased risk for development of periodontitis was also reported.8 Increased expression of NOD1 and NOD2 in the epithelial cells and inflammatory cells of humans in both healthy and diseased gingiva with no significant difference was reported among various periodontal conditions. 9 A remarkable reduction in alveolar bone resorption and osteoclastogenesis was seen in a study on mice carried out in preclinical settings that suggested the possibility of NOD2 triggering periodontal bone loss.10

NLRP1 inflammasome

The first ever NLR described to form an inflammasome complex was NLRP1. It is encoded by a single gene in humans that contains a PYD (pyrin) domain, FIIND (function-to-find domain), and CARD domain. 11, 12 Recent research has shown that NLRP1 signalling is induced by the peptidoglycan component muramyl dipeptide (MDP) of bacterial cell walls and anthrax lethal factor (LF) of the Bacillus anthracis lethal toxin (LeTx). 12, 13 The toxin consists of protective antigen (PA) and lethal factor (LF). These two proteins create pores in the host cell membrane and subsequently activates NLRP1β14 causing inflammasome assembly.

It was believed earlier that the human NLRP1 comprised of NLRP1, ASC, caspase-1, and caspase-5. Processing of caspase-5 is stimulated by the interaction between NLRP1 and caspase-5 but the adaptor protein ASC is required to mediate the interaction and to subsequently process caspase-1.11 Despite all the observations made, the specific triggers of human NLRP1 and murine NLRP1a/c still remain to be discovered. It is even possible that they may have different roles in mouse and man as some reports showed that ATP can bind to NLRP1 thus activating it.11 ATP binding was inhibited by the anti-apoptotic proteins BCL-2 and BCL-xl through inhibition of NLRP1 inflammasome activation.5

Interestingly, a role for NOD2 (NLRC2) in the NLRP1 inflammasome assembly in response to both MDP and anthrax lethal toxin was suggested.15 NOD2 is a sensor for MDP and initiates activation of NF-κB and mitogen activated protein kinase (MAPK) via an adaptor protein RIP2 (receptor-interacting-serine/threonine-protein kinase 2).16 In a cell-free system, NLRP1 can activate caspase-1 in response to MDP but to detect both MDP and LeTx in vivo, NOD2 is required. NOD2 seemed to generate signals necessary for caspase activation, pro-IL1β expression and secretion of mature IL-1β.15, 17 However, further research is required to comprehend the exact function of NLRP1 and NOD2 in response to anthrax lethal toxin and MDP. The risk for developing several autoimmune diseases, such as generalized vitiligo, vitiligo-associated type I diabetes, and Addison’s disease18, 19 rheumatoid arthritis, systemic sclerosis, and Crohn’s disease were attributed to single nucleotide polymorphisms in the human genomic NLRP1 region.20 The participation of NLRP1a in pyroptosis of hematopoietic cells were also observed due to mutations resulting in hyperactive inflammasomes and IL-1β release.21 However, the specific stimuli involved in regulation of NLRP1a inflammasome are elusive and only further research in the area can define the molecular mechanisms behind it.

Decreased NLRP1 expression was observed in the samples of healthy, chronic and aggressive periodontitis gingiva with more frequent expression in the epithelium and connective tissue of individuals with aggressive periodontitis. Most studies attempting to explore its role have concentrated on autoinflammatory diseases, with little information on it in the context of periodontal diseases.

NLRP3 inflammasome

NLRP3 is undoubtedly the most extensively studied NLR. Its expression is low in myeloid cells and Toll-like receptor agonists, such as lipopolysaccharide (LPS), and inflammatory cytokines, such as TNF-α transcriptionally induces it.5 The involvement of NLRP3 has been detected in a considerable number of inflammatory diseases like diabetes mellitus, obesity and atherosclerosis. As we understand, NLRP3 activation is basically a two-step process and three models pertaining to its activation were put forward which include: the ion flux model, the reactive oxygen species (ROS) model, and the lysosome rupture model.22 In the ion flux model, there were changes in cytosolic levels of K+, Ca2+, and H+. Extracellular ATP induces rapid K+ efflux via activation of the ATP-gated ion channel P2X7;23 Nigericin creates a K+ pore in the cell membrane; the influenza M2 protein triggers export of H+ ions from the Golgi complex into the cytosol; and high concentrations of extracellular Ca2+, increase cytosolic Ca2+, and cAMP (cyclic adenosine monophosphate).22

The generation of reactive oxygen species (ROS) by many NLRP3-activating stimuli, like adenosine triphosphate (ATP), alum, uric acid, and Nigericin1 has been observed in NLRP3 activation although its exact function here is disputable. Some studies described an alternative role for ROS in inflammasome activation where oxidized mitochondrial DNA released from dysfunctional mitochondria can directly activate NLRP3.24 Recently, NIMA(never in mitosis gene a)-related kinase 7 (NEK7), a serine-threonine kinase was also observed to induce NLRP3 activation. 25, 26 There is still no clarity regarding NEK7 binding and NLRP3 oligomerization, although it is required for ASC polymerization.27 Another recently observed regulator of NLRP3 activation was PKD (protein kinase D).28

The non-canonical pathway of NLRP3 activation was more complex29 than the canonical pathway that activates NLRP3 directly. The noncanonical pathway involves activation of caspase-11 by intracellular LPS30 but the exact mechanism here still remains to be understood. Nevertheless, currently there is no agreement on a standard mechanism explaining NLRP3 activation due to the diverse range of stimuli involved here. Not many clinical studies have explored the relation of NLRP3 with periodontal disease. A four to five fold increase in NLRP3 mRNA expression was reported in gingival tissues of individuals with chronic periodontitis with almost a seven fold increase in case of aggressive periodontitis.31, 32 Immunohistochemistry revealed that the increased expression was more pronounced in the epithelial layer, thus suggesting the role of NLRP3 in epithelium as part of the host’s innate immunity in the gingival sulcus and tissues.31 Even inflammation confined to gingiva or that which has not reached the bone increased NLRP3 levels. 32 Salivary samples of individuals with chronic and aggressive periodontitis also reported remarkably increased expressions of NLRP3.33 Moreover, a positive correlation between mRNA expressions of NLRP3 and IL1β and IL18 in gingival tissues was reported.32 Nevertheless, no study has described a genetic background for NLRP3 with regard to periodontal disease as yet.

NLRP6

As opposed to the case of NLRP3, non-hematopoietic cells like epithelial cells and goblet cells in the intestine show an increased expression of NLRP6,34 although there are some who claim that its expression is mainly observed in hematopoietic cells.35 It has been shown to cause strong proinflammatory effects in NLRP6-deficient mice in response to bacterial challenge due to negative regulation of NF-kB and MAPK signalling pathways.36 Macrophages infected with Listeria monocytogenes, E. coli, and Salmonella also demonstrated similar processes.35 With this background, the role of NLRP6 in caspase-1 activation at mucosal sites was believed to be of utmost importance.34 It was demonstrated that subsequent IL-18 release promoted the production of antimicrobial peptides which act against microbial dysbiosis.37 The identity of the ligand activating NLRP6 remained elusive until recently taurine, a microbial metabolite was described.38 Mucosal immunity in the gut has been attributed to the production of a specific metabolite profile enriched in taurine by the commensal non-pathogenic microbiota that binds to NLRP6.38 All these data imply its key role in gut homeostasis and associated disorders.

Furthermore, in the context of viral infection, it was demonstrated that systemic infections with encephalomyocarditis and norovirus caused NLRP6 to affect viral loads at mucosal sites specifically in the gastrointestinal tract.39 It was also reported that oral infections with the same viruses needed NLRP6-mediated defense to survive.40 However, adequate research has not been conducted to establish its importance at nonmucosal sites.

NLRP12

NLRP12 is similar to NLRP6 in many ways. NLRP12 also has shown to be involved in the regulation of NF-kB and MAPK pathways to maintain intestinal homeostasis. 41 In addition, it was observed that after Yersinia pestis infection, IL-18 and IL-1β production was regulated by this inflammasome and that NLRP12-deficient mice were more vulnerable to bacterial challenge. 42 Nevertheless, the specific triggers for NLPR12 activation and the precise mechanisms involved still remains unknown as is the case with NLRP6.

NLRC4 inflammasome

NLRC4 (also known as IPAF, Card12) is found to be involved in infections with various gram-negative bacteria. It contains an N-terminal CARD, a central NBD, and a C-terminal LRR domain.5 NLRC4 activation, like other NLRs involves the deletion of the LRR (leucine rich repeat) domain. Sometimes CARD – CARD interactions were observed in the association of NLRC4 specifically with the CARD domain of pro-caspase-1.43 However, it is noteworthy that there are other studies demonstrating that NLRC4-mediated caspase-1 activation needs the functional involvement of ASC. Hence, the requirement of ASC here is disputable. It maybe that ASC stabilizes or facilitates caspase-1 recruitment to NLRC4. Alternatively, NLRC4 may also recruit ASC via PYD–PYD interaction by cooperating with a PYD-containing NLRP protein to activate caspase-1.5

NLRC4 recognizes the following components of pathogenic bacteria: 44

  1. Flagellin, the building block of their locomotion apparatus, and

  2. Proteins from the type III and IV bacterial secretion systems known to inject virulence factors into the host cell.

The flagella which is a structure on the bacterial cell wall that allows bacterial motility contain polymers of the protein flagellin which are its structural compounds. Flagellin is an apt example of a PAMP as it is an evolutionarily conserved element that aids in bacterial motility. NLRC4-deficient macrophages show a significant reduction in caspase-1 activation, release of IL-1β, and pyroptosis following infection with the gram-negative Salmonella typhimurium,45 Legionella pneumophila,46 Pseudomonas aeruginosa, and Shigella flexneri.45 These bacteria share either a type III (T3SS) or type IV secretion system. An impaired ability of S. typhimurium and L. pneumophila strains deficient in flagellin to activate caspase-1 in macrophages were initially observed.46 Even the delivery of purified flagellin into the host cell activated caspase-1 in an NLRC4-dependent manner. Notably, caspase-1 activation by NLRC4 is independent of TLR5.47 These results led many to hypothesize that flagellin is the primary PAMP activating NLRC4. However, it was observed that only flagellin from certain bacteria activated NLRC4. Even if delivered to the host cytosol, flagellin of E. coli does not activate inflammasomes.48 On the other hand, the flagellated gram-positive Listeria monocytogenes, was shown to trigger NLRC4 activation, even though they lack a secretion system. This provides flagellin access to the cytosol and thus NLRC4 and Listeria escapes the phagolysosome to multiply in the cytosol.49 On the contrary, the nonflagellated S. flexneri was shown to activate caspase-1 in an NLRC4-dependent manner. 5

It was demonstrated recently that phosphorylation of NLRC4 was necessary for inflammasome assembly.22 Surprisingly, intraperitoneal delivery of flagellin in mouse activated NLRC4 which induced host mortality within 30 minutes. It is quite interesting to note that the inflammasome- dependent response here is determined by the production of eicosanoids and not IL-1β or IL-18. This is followed by a rapid loss of vascular fluid and mortality.50 The mechanism governing this response is currently unknown but it highlights its relevance in cellular responses.

ALR family

The ALR family has four members in humans out of which IFI16 (Interferon Gamma Inducible Protein 16) and AIM2 (absent in melanoma 2) are the only inflammasome-forming members. While AIM2 identifies only double-stranded DNA, IFI16 is able to identify both single-stranded and double-stranded DNA of various viruses and intracellular bacteria. They are characterized by an N-terminal PYD and a C-terminal hematopoietic interferon-inducible nuclear protein with 200–amino acid repeat (HIN200) domain. In AIM2, the PYD is capable of interacting with the PYD of ASC, unlike other ALRs.39 This specific interaction recognises AIM2 as the only member of the family capable of forming an inflammasome. The human ALR, γ-IFN–inducible protein 16, is considered a putative inflammasome.39

AIM2-Like Receptors

AIM2 (absent in melanoma 2) was originally identified as a γ-IFN–inducible protein in a tumor suppressor screen. Later on, it was shown to detect nucleic acids that played a role in assembling inflammasomes, especially double-stranded DNA which could either be of bacterial/viral origin or self DNA from apoptotic cells.28, 51 AIM2 and the ALRs belong to the PYHIN family, which comprises of proteins that have a PYRIN domain and a hematopoietic IFN-inducible nuclear protein with 200-amino acids (HIN-200) domain.6 Structural analysis also attributed an autoinhibitory role to the HIN200 domain due to its interaction with the PYD of AIM2 in the absence of a ligand.

Although AIM2 binds to both viral and bacterial DNA, there is a remarkable difference in the mechanisms involved in each case. Type I IFN signalling is involved in AIM2 activation induced by bacteria such as Francisella, but not viruses like mouse cytomegalovirus.52 Be that as it may, not all bacteria and viruses activate AIM2 inflammasome. Recent studies point towards certain bacteria, such as Mycobacterium and Legionella, that can reduce dsDNA release and thereby avoid detection by AIM2.53 Recently a link between the pathology associated with chemotherapeutic drug therapy and AIM2 without affecting their anti-cancer properties was observed.54 Various autoinflammatory diseases, such as psoriasis, systemic lupus erythematosus, and abdominal aortic aneurysm55 have been associated with AIM2. Therefore, all these observations raise the possibility of AIM2 being a therapeutic target for these autoimmune disorders in future.

Another member of the HIN200 family, p202, lacks the N-terminal PYD and may negatively regulate AIM2 inflammasome.56 In addition to AIM2, humans have three more ALRs, IFI16, IFIX (interferon-inducible protein X ), and MNDA (myeloid cell nuclear differentiation antigen), whereas mice express an extended range of ALRs consisting of at least 13 members.57 Although most of these are poorly characterized, many murine ALRs were reported to have triggered IL-1β production, suggesting their role in inflammasome assembly.57

IFI16 and RIG-I-Like Receptors

The ability of human IFI16 to form an inflammasome in response to Kaposi’s sarcoma–associated herpes virus infection, and activation of IFI16 in quiescent CD4+ T cells during HIV infection was found to trigger extensive pyroptosis of T cells. Hence, it is observed to be a significant contributor of depletion of CD4+ T cells during progression to AIDS.58 Some of IFI16's are even capable of modulating AIM2 either through its upregulation or down regulation.59 Finally, the RIG-I like receptor (RLR) family member RIG-I, which is best known to stimulate type I IFN production on recognition of viral RNA, was also found to be capable of forming an inflammasome.22 However, it still remains to be clear as to what exactly determines if RIG-I should form an inflammasome or if it should just trigger type I IFN production. Although the expression of IFI16 was higher in inflammatory bowel diseases like Crohn's disease and ulcerative colitis, the exact function of IFI16 and AIM2 in periodontal disease pathogenesis remains elusive. In periodontitis models, its variations in tissue expression were observed depending on periodontal disease status, limited tissue characterization and correlation of specific gene variants.60 AIM2 levels were almost two times higher in the lamina propria and epithelium of chronic periodontitis and aggressive periodontitis cases.31

Current research indicates an upregulation of IFI16 and AIM2 in diseased tissues which may reflect a disruption of the epithelial barrier function. Any variants of these proteins seemed to affect their expression or function thus, predisposing an individual to periodontal disease by bringing about changes in the oral microbial composition.

Therapeutics targeting the inflammasome

The host response has been long established as a major factor in contributing to periodontal destruction through inflammation and disruption of tissue homeostasis. Most of the treatment approaches to improve periodontal status of an individual focused on antimicrobial therapy rather than modulation of host response. An uncontrolled inflammasome activity results in pro-inflammatory effects contributing to the development of many diseases. Therefore, approaches involving inhibitors/antagonists targeting inflammasome components, their activation status and cytokine production are important in treating periodontal diseases.

Inhibiting the upstream intracellular signaling pathways

  1. Many upstream signals like ion flux, lysosomal disruption, mitochondrial damage and ROS can cause NLRP3 activation. A drug called Allopurinol is considered a standard treatment of hyperuricemia associated with treatment of gout (a form of arthritis that is NLRP3 inflammasome dependent) 61 and kidney stones. 62

  2. The therapeutic drug SS-31 selectively targets cardiolipin, a phospholipid in the inner mitochondrial membrane. 63 This prevents excessive ROS production. However, no studies have evaluated the effects of these drugs in periodontal diseases in a preclinical or clinical setting so far.

Blocking inflammasome components

  1. IL-1β and IL-18 release is preceded by caspase-1 activation. Three drugs targeting caspase-1, Emricasan, VX-740 and VX-765, have been tested in humans.64 The micro-computed tomography results here showed that caspase-1 inhibition caused a 50% reduction of alveolar bone loss than normal.

  2. Bruton's tyrosine kinase (BTK) is a non-receptor tyrosine kinase, best known for its important role during B cell development. During NLRP3 activation, it communicates with NLRP3 and ASC. 65 The BTK inhibitor ibrutinib (PCI-32765, Imbruvica) is a FDA-approved drug for the treatment of various B cell cancers. Recently, another BTK inhibitor, acalabrutinib (ACP-196), has shown to prevent alveolar bone loss 66 indicating its possible role in treating periodontal diseases.

  3. Recently, it was observed that interleukin-1β levels reduced to almost half in experimental periodontitis67 during colchicine treatment.

Inhibiting inflammasome-mediated cytokines

  1. Interleukin-1β is the most potent pro-inflammatory cytokine released by activation of inflammasomes and is implicated in many inflammasome-associated diseases and plays a critical role in the periodontal disease progression. 68 eg: monoclonal antibody targeting IL-1β (canakinumab), by a modified IL-1β receptor antagonist (anakinra) or by a soluble decoy receptor (rilonacept). A combination of IL-1 inhibitor and TNFα inhibitor in a study, demonstrated a 50% reduction in the levels of radiographic bone loss compared to the control sites 69 suggesting that both IL-1β and TNF-α may act as potential targets for treating periodontal disease.

  2. GSK1070806 is an anti-IL-18 monoclonal antibody. It was studied in a diabetic population where no improvements in glucose control were observed. 70 Nevertheless, further research is required to evaluate these therapeutics targeting IL-18 in the context of periodontal diseases.

Conclusion

This is a brief review explaining the functions and activation mechanisms of inflammasomes illustrating their increasingly significant roles in infectious, autoimmune, and autoinflammatory diseases. While some of them have been established for years, further research is paramount to elucidate many of the unique post-translational regulatory mechanisms that modulate their activation, repression and susceptibility to various infections The available evidence indicate that various inflammasome components are expressed in higher levels in saliva, gingival crevicular fluid and periodontal tissues. In light of the observations made so far, it is possible that individuals with autoinflammatory diseases may have certain unknown biological pathways that may lead to periodontal disease. Furthermore, the pathways involved in the regulation of each of these inflammasomes may present new targets for drugs, thereby resulting in minimal pathological effects in many diseases.

Source of Funding

No financial support was received for the work within this manuscript.

Conflict of Interest

The authors declare they have no conflict of interest.

References

1 

K Schroder J Tschopp The InflammasomesCell20101408213210.1016/j.cell.2010.01.040

2 

SM Man T-D Kanneganti Converging roles of caspases in inflammasome activation, cell death and innate immunityNat Rev Immunol201616172110.1038/nri.2015.7

3 

R Heilig MS Dick L Sborgi E Meunier S Hiller P Broz The Gasdermin-D pore acts as a conduit for IL-1β secretion in miceEur J Immunol20184845849210.1002/eji.201747404

4 

JP Ting RC Lovering ES Alnemri J Bertin JM Boss BK Davis The NLR Gene Family: A Standard NomenclatureImmun2008283285710.1016/j.immuni.2008.02.005

5 

F Bauernfeind A Ablasser E Bartok S Kim J Schmid-Burgk T Cavlar Inflammasomes: current understanding and open questionsCell Mol Life Sci2011687658310.1007/s00018-010-0567-4

6 

SA Schattgen KA Fitzgerald The PYHIN protein family as mediators of host defensesImmunological Rev201124311091810.1111/j.1600-065x.2011.01053.x

7 

RC Ireton M Jr. Gale RIG-I Like Receptors in Antiviral Immunity and Therapeutic ApplicationsViruses2011369061910.3390/v3060906

8 

SN Papageorgiou M Hagner AVB Nogueira A Franke A Jäger J Deschner Inflammatory bowel disease and oral health: systematic review and a meta-analysisJ Clin Periodontol20174443829310.1111/jcpe.12698

9 

Y. Sugawara A. Uehara Y. Fujimoto S. Kusumoto K. Fukase K. Shibata Toll-like Receptors, NOD1, and NOD2 in Oral Epithelial CellsJ Dent Res2006856524910.1177/154405910608500609

10 

J A Souza M C Medeiros F R Rocha Role of NOD2 and RIP2 in host-microbe interactions with Gram-negative bacteria: insights from the periodontal disease modelInnate Immun2016228598611

11 

F Martinon K Burns J Tschopp The InflammasomeMol Cell2002104172610.1016/s1097-2765(02)00599-3

12 

ED Boyden WF Dietrich Nalp1b controls mouse macrophage susceptibility to anthrax lethal toxinNat Genet2006382240410.1038/ng1724

13 

B Faustin L Lartigue J-M Bruey F Luciano E Sergienko B Bailly-Maitre Reconstituted NALP1 Inflammasome Reveals Two-Step Mechanism of Caspase-1 ActivationMol Cell2007257132410.1016/j.molcel.2007.01.032

14 

J Chavarría-Smith RE Vance Direct Proteolytic Cleavage of NLRP1B Is Necessary and Sufficient for Inflammasome Activation by Anthrax Lethal FactorPLoS Pathog201396e100345210.1371/journal.ppat.1003452

15 

L.-C. Hsu S. R. Ali S. McGillivray P.-H. Tseng S. Mariathasan E. W. Humke A NOD2-NALP1 complex mediates caspase-1-dependent IL-1  secretion in response to Bacillus anthracis infection and muramyl dipeptideProc National Acad Sci2008105227803810.1073/pnas.0802726105

16 

SE Girardin IG Boneca J Viala M Chamaillard A Labigne G Thomas Nod2 Is a General Sensor of Peptidoglycan through Muramyl Dipeptide (MDP) DetectionJ Biol Chem20032781188697210.1074/jbc.c200651200

17 

G Ferwerda M Kramer D de Jong A Piccini LA Joosten I DevesaGiner Engagement of NOD2 has a dual effect on proIL-1β mRNA transcription and secretion of bioactive IL-1βEur J Immunol20083811849110.1002/eji.200737103

18 

Y Jin SA Birlea PR Fain RA Spritz Genetic Variations in NALP1 Are Associated with Generalized Vitiligo in a Romanian PopulationJ Investig Dermatol20071271125586210.1038/sj.jid.5700953

19 

Y Jin CM Mailloux K Gowan SL Riccardi G LaBerge DC Bennett NALP1in Vitiligo-Associated Multiple Autoimmune DiseaseN Eng J Med20073561212162510.1056/nejmoa061592

20 

JN Finger JD Lich LC Dare MN Cook KK Brown C Duraiswami Autolytic Proteolysis within the Function to Find Domain (FIIND) Is Required for NLRP1 Inflammasome ActivityJ Biol Chem20122873025030710.1074/jbc.m112.378323

21 

SL Masters M Gerlic D Metcalf S Preston M Pellegrini JA O’Donnell NLRP1 Inflammasome Activation Induces Pyroptosis of Hematopoietic Progenitor CellsImmun201237610092310.1016/j.immuni.2012.08.027

22 

M. R. de Zoete N. W. Palm S. Zhu R. A. Flavell InflammasomesCold Spring Harbor Perspect Biol20146a01628710.1101/cshperspect.a016287

23 

V Pétrilli S Papin C Dostert A Mayor F Martinon J Tschopp Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentrationCell Death Differ20071491583910.1038/sj.cdd.4402195

24 

K Shimada T R Crother J Karlin J Dagvadorj N Chiba S Chen Oxidized Mitochondrial DNA Activates the NLRP3 Inflammasome during ApoptosisImmun20123634011410.1016/j.immuni.2012.01.009

25 

JL Schmid-Burgk D Chauhan T Schmidt TS Ebert J Reinhardt E Endl A Genome-wide CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) Screen Identifies NEK7 as an Essential Component of NLRP3 Inflammasome ActivationJ Biol Chem20162911103910.1074/jbc.c115.700492

26 

H Shi Y Wang X Li X Zhan M Tang M Fina NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome componentNat immunol20161732508

27 

JP Green S Yu F Martín-Sánchez P Pelegrin G Lopez-Castejon CB Lawrence Chloride regulates dynamic NLRP3-dependent ASC oligomerization and inflammasome primingProc National Acad Sci201811540E93718010.1073/pnas.1812744115

28 

Z Zhang G Meszaros W He Y Xu H de Fatima Magliarelli L Mailly Protein kinase D at the Golgi controls NLRP3 inflammasome activationJ Exp Med2017214926719310.1084/jem.20162040

29 

N Kayagaki S Warming M Lamkanfi LV Walle S Louie J Dong Non-canonical inflammasome activation targets caspase-11Nat201147973711172110.1038/nature10558

30 

N. Kayagaki M. T. Wong I. B. Stowe S. R. Ramani L. C. Gonzalez S. Akashi-Takamura Noncanonical Inflammasome Activation by Intracellular LPS Independent of TLR4Sci201334161511246910.1126/science.1240248

31 

F Xue R Shu Y Xie The expression of NLRP3, NLRP1 and AIM2 in the gingival tissue of periodontitis patients: RT-PCR study and immunohistochemistryArch Oral Biol20156069485810.1016/j.archoralbio.2015.03.005

32 

N. Bostanci G. Emingil B. Saygan O. Turkoglu G. Atilla M. A. Curtis Expression and regulation of the NALP3 inflammasome complex in periodontal diseasesClin Exp Immunol200915734152210.1111/j.1365-2249.2009.03972.x

33 

Diana M. Isaza-Guzmán Verónica M. Medina-Piedrahíta Carolina Gutiérrez-Henao Sergio I. Tobón-Arroyave Salivary Levels of NLRP3 Inflammasome-Related Proteins as Potential Biomarkers of Periodontal Clinical StatusJournal of Periodontology20178812132913380022-3492, 1943-367010.1902/jop.2017.170244Wileyhttps://dx.doi.org/10.1902/jop.2017.170244

34 

M Wlodarska CA Thaiss R Nowarski J Henao-Mejia J-P Zhang EM Brown NLRP6 Inflammasome Orchestrates the Colonic Host-Microbial Interface by Regulating Goblet Cell Mucus SecretionCell2014156510455910.1016/j.cell.2014.01.026

35 

GY Chen M Liu F Wang J Bertin G Núñez A Functional Role for Nlrp6 in Intestinal Inflammation and TumorigenesisJ Immunol201118671879410.4049/jimmunol.1100412

36 

PK Anand T-D Kanneganti NLRP6 in infection and inflammationMicrobes Infect201315661810.1016/j.micinf.2013.06.009

37 

E Elinav T Strowig AL Kau J H-Mejia CA Thaiss CJ Booth NLRP6 Inflammasome Regulates Colonic Microbial Ecology and Risk for ColitisCell201114557455710.1016/j.cell.2011.04.022

38 

M Levy CA Thaiss D Zeevi L Dohnalová GZ-Schapira JA Mahdi Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome SignalingCell2015163614284310.1016/j.cell.2015.10.048

39 

D Sharma T-D Kanneganti The cell biology of inflammasomes: Mechanisms of inflammasome activation and regulationJ Cell Biol201621366172910.1083/jcb.201602089

40 

P Wang S Zhu L Yang S Cui W Pan R Jackson Nlrp6 regulates intestinal antiviral innate immunitySci2015350626282630

41 

IC Allen JE Wilson M Schneider JD Lich RA Roberts JC Arthur NLRP12 Suppresses Colon Inflammation and Tumorigenesis through the Negative Regulation of Noncanonical NF-κB SignalingImmun20123657425410.1016/j.immuni.2012.03.012

42 

GI Vladimer D Weng SW Montminy Paquette S Kailasan Vanaja VAK Rathinam MH Aune The NLRP12 Inflammasome Recognizes Yersinia pestisImmun20123719610710.1016/j.immuni.2012.07.006

43 

JL Poyet SM Srinivasula M Tnani M Razmara T Fernandes-Alnemri ES Alnemri Identification of Ipaf, a Human Caspase-1-activating Protein Related to Apaf-1J Biol Chem200127630283091310.1074/jbc.c100250200

44 

E. A. Miao D. P. Mao N. Yudkovsky R. Bonneau C. G. Lorang S. E. Warren Innate immune detection of the type III secretion apparatus through the NLRC4 inflammasomeProc National Acad Sci2010107730768010.1073/pnas.0913087107

45 

S Mariathasan K Newton DM Monack D Vucic DM French WP Lee Differential activation of the inflammasome by caspase-1 adaptors ASC and IpafNat20044306996213810.1038/nature02664

46 

DS Zamboni KS Kobayashi T Kohlsdorf Y Ogura EM Long RE Vance The Birc1e cytosolic pattern-recognition receptor contributes to the detection and control of Legionella pneumophila infectionNat Immunol2006733182510.1038/ni1305

47 

L Franchi A Amer M Body-Malapel T-D Kanneganti N Özören R Jagirdar Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1β in salmonella-infected macrophagesNat Immunol2006765768210.1038/ni1346

48 

T Ren DS Zamboni CR Roy WF Dietrich RE Vance Flagellin-Deficient Legionella Mutants Evade Caspase-1- and Naip5-Mediated Macrophage ImmunityPLoS Pathog200623e1810.1371/journal.ppat.0020018

49 

SE Warren DP Mao AE Rodriguez EA Miao A Aderem Multiple Nod-Like Receptors Activate Caspase 1 during Listeria monocytogenes InfectionJ Immunol20081801175586410.4049/jimmunol.180.11.7558

50 

Jvon Moltke NJ Trinidad M Moayeri AF Kintzer SB Wang N van Rooijen Rapid induction of inflammatory lipid mediators by the inflammasome in vivoNat201249074181071110.1038/nature11351

51 

Tilmann Bürckstümmer Christoph Baumann Stephan Blüml Evelyn Dixit Gerhard Dürnberger Hannah Jahn Melanie Planyavsky Martin Bilban Jacques Colinge Keiryn L Bennett Giulio Superti-Furga An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasomeNature Immunology20091032662721529-2908, 1529-291610.1038/ni.1702Springer Science and Business Media LLChttps://dx.doi.org/10.1038/ni.1702

52 

SM Man R Karki RKS Malireddi G Neale P Vogel M Yamamoto The transcription factor IRF1 and guanylate-binding proteins target activation of the AIM2 inflammasome by Francisella infectionNat Immunol20151654677510.1038/ni.3118

53 

S Shah A Bohsali SE Ahlbrand L Srinivasan VAK Rathinam SN Vogel Cutting Edge: Mycobacterium tuberculosis but Not Nonvirulent Mycobacteria Inhibits IFN-β and AIM2 Inflammasome–Dependent IL-1β Production via Its ESX-1 Secretion SystemJ Immunol201319173514810.4049/jimmunol.1301331

54 

DE Place TD Kanneganti Recent advances in inflammasome biologyCurr Opin Immunol20185032810.1016/j.coi.2017.10.011

55 

S Dihlmann P Erhart A Mehrabi A Nickkholgh F Lasitschka D Böckler Increased Expression and Activation of Absent in Melanoma 2 Inflammasome Components in Lymphocytic Infiltrates of Abdominal Aortic AneurysmsMol Med2014201230710.2119/molmed.2013.00162

56 

T. L. Roberts A. Idris J. A. Dunn G. M. Kelly C. M. Burnton S. Hodgson HIN-200 Proteins Regulate Caspase Activation in Response to Foreign Cytoplasmic DNASci2009323591710576010.1126/science.1169841

57 

RL Brunette JM Young DG Whitley IE Brodsky HS Malik DB Stetson Extensive evolutionary and functional diversity among mammalian AIM2-like receptorsJ Exp Med20122091119698310.1084/jem.20121960

58 

K. M. Monroe Z. Yang J. R. Johnson X. Geng G. Doitsh N. J. Krogan IFI16 DNA Sensor Is Required for Death of Lymphoid CD4 T Cells Abortively Infected with HIVSci201434361694283210.1126/science.1243640

59 

P H Wang Z W Ye J J Deng K L Siu W W Gao V Chaudhary Inhibition of AIM 2 inflammasome activation by a novel transcript isoform of IFI 16EMBO Rep2018191045737

60 

S Offenbacher K Divaris SP Barros KL Moss JT Marchesan T Morelli Genome-wide association study of biologically informed periodontal complex traits offers novel insights into the genetic basis of periodontal diseaseHuman Mol Genet2016251021132910.1093/hmg/ddw069

61 

AK So F Martinon Inflammation in gout: mechanisms and therapeutic targetsNat Rev Rheumatol201713116394710.1038/nrrheum.2017.155

62 

P Pacher A Nivorozhkin C Szabó Therapeutic Effects of Xanthine Oxidase Inhibitors: Renaissance Half a Century after the Discovery of AllopurinolPharmacol Rev20065818711410.1124/pr.58.1.6

63 

J T Marchesan M S Girnary K Moss E T Monaghan G J Egnatz Y Jiao Role of inflammasomes in the pathogenesis of periodontal disease and therapeuticsPeriodontol200082193114

64 

S Cornelis K Kersse N Festjens M Lamkanfi P Vandenabeele Inflammatory Caspases: Targets for Novel TherapiesCurr Pharm Des20071343678510.2174/138161207780163006

65 

M Ito T Shichita M Okada R Komine Y Noguchi A Yoshimura Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injuryNat Commun201561736010.1038/ncomms8360

66 

N K Pokhrel Y G Kim H J Kim A novel Bruton's tyrosine kinase inhibitor acalabrutinib suppresses osteoclast differentiation and P. gingivalis lipopolysaccharide-induced alveolar bone resorptionJ Periodontol201819

67 

CA Aral K Aral A Yay Ö Özçoban A Berdeli R Saraymen Effects of colchicine on gingival inflammation, apoptosis, and alveolar bone loss in experimental periodontitisJ Periodontol20188955778510.1002/jper.17-0359

68 

AA Jesus R Goldbach-Mansky IL-1 Blockade in Autoinflammatory SyndromesAnn Rev Med20146512234410.1146/annurev-med-061512-150641

69 

T. W. Oates D. T. Graves D. L. Cochran Clinical, radiographic and biochemical assessment of IL-1/TNF-α antagonist inhibition of bone loss in experimental periodontitisJ Clin Periodontol20022921374310.1034/j.1600-051x.2002.290208.x

70 

EA McKie JL Reid PC Mistry SL DeWall L Abberley PD Ambery A Study to Investigate the Efficacy and Safety of an Anti-Interleukin-18 Monoclonal Antibody in the Treatment of Type 2 Diabetes MellitusPLOS ONE2016113e015001810.1371/journal.pone.0150018



jats-html.xsl


This is an Open Access (OA) journal, and articles are distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as appropriate credit is given and the new creations are licensed under the identical terms.

  • Article highlights
  • Article tables
  • Article images

View Article

PDF File   Full Text Article


Copyright permission

Get article permission for commercial use

Downlaod

PDF File   XML File   ePub File


Digital Object Identifier (DOI)

Article DOI

https://doi.org/10.18231/j.ijpi.2020.036


Article Metrics






Article Access statistics

Viewed: 1746

PDF Downloaded: 968



Medical Abbreviation List