Next Article in Journal
A Beckwith–Wiedemann-Associated CDKN1C Mutation Allows the Identification of a Novel Nuclear Localization Signal in Human p57Kip2
Next Article in Special Issue
Inflammation Is Present, Persistent and More Sensitive to Proinflammatory Triggers in Celiac Disease Enterocytes
Previous Article in Journal
Structural Evaluation of the Spike Glycoprotein Variants on SARS-CoV-2 Transmission and Immune Evasion
Previous Article in Special Issue
Pediatric Celiac Disease Patients Show Alterations of Dendritic Cell Shape and Actin Rearrangement
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Programmed Cell Death in the Small Intestine: Implications for the Pathogenesis of Celiac Disease

Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata B1900, Argentina
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(14), 7426; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147426
Submission received: 29 May 2021 / Revised: 17 June 2021 / Accepted: 18 June 2021 / Published: 10 July 2021
(This article belongs to the Special Issue Pro-inflammatory Nutrient: Focus on Gliadin and Celiac Disease)

Abstract

:
The small intestine has a high rate of cell turnover under homeostatic conditions, and this increases further in response to infection or damage. Epithelial cells mostly die by apoptosis, but recent studies indicate that this may also involve pro-inflammatory pathways of programmed cell death, such as pyroptosis and necroptosis. Celiac disease (CD), the most prevalent immune-based enteropathy, is caused by loss of oral tolerance to peptides derived from wheat, rye, and barley in genetically predisposed individuals. Although cytotoxic cells and gluten-specific CD4+ Th1 cells are the central players in the pathology, inflammatory pathways induced by cell death may participate in driving and sustaining the disease through the release of alarmins. In this review, we summarize the recent literature addressing the role of programmed cell death pathways in the small intestine, describing how these mechanisms may contribute to CD and discussing their potential implications.

1. Introduction

1.1. Programmed Cell Death (PCD)

Many new cell death pathways have been discovered in recent years. From the original concept of apoptosis as a unique form of immunologically silent programmed cell death, to proinflammatory necrosis, a broad spectrum of different pathways is known to develop in specific conditions. Apoptosis is characterized by cell shrinkage and chromatin condensation, followed by fragmentation of the entire cell into small apoptotic bodies, which are cleared away by macrophages without initiating an inflammatory response. Unlike apoptosis, other forms of PCD are not immunologically silent and are involved in driving and maintaining a variety of metabolic and inflammatory disorders. These pathways include necroptosis, pyroptosis and ferroptosis and they can result in the release of proinflammatory molecules such as alarmins (IL-33, HMGB1, IL-1α) and proinflammatory cytokines (IL-1β and IL-18) [1,2,3]. Importantly, inflammatory PCD enables the release of molecules such as IL-1β without cell death occurring [4]. Studying the molecular pathways involved in these processes is important in order to gain insight into the pathogenesis of inflammatory disorders and for the development of therapeutic interventions.

1.2. Celiac Disease: A Complex Small Intestine Pathology

1.2.1. Antigen-Specific CD4+ T Cells

Celiac disease (CD) is a highly prevalent chronic inflammatory enteropathy which occurs in genetically susceptible individuals as a consequence of an immune response to gluten proteins derived from wheat, barley and rye [5,6]. It affects the proximal small intestine, leading to villus atrophy and crypt hyperplasia, together with increased numbers of lamina propria and intraepithelial lymphocytes (IELs). However, the histological changes and clinical presentation are variable, and many cases remain asymptomatic, leading to a large number of undiagnosed patients [7]. The histological changes are caused by an immune response to dietary gluten in the small intestine mucosa. As gluten peptides are resistant to proteolysis by gastrointestinal enzymes, long peptides remain in the lumen of the intestine. After crossing the epithelial layer [8], some of these peptides are deamidated by the enzymatic activity of transglutaminase 2 (TG2), generating epitopes with the ability to bind to the disease specifying MHC molecules HLA-DQ2 and DQ8. In the lamina propria, native and deamidated gluten derived peptides are taken up and presented in HLA molecules by dendritic cells (DC). Based on studies in mice, it is believed that lamina propria DC migrate to the draining mesenteric lymph nodes where they encounter naïve, antigen-specific CD4+ T cells and induce their differentiation into Th1 CD4+ T cells, which migrate back to the lamina propria [9]. However, this has not been shown directly in humans with CD. In the lamina propria, gluten-specific CD4+ cells produce IFNγ, the dominant cytokine in the chronic inflammatory process [6,10,11].

1.2.2. Cytotoxic Mechanisms

Several gluten peptides have been found that can bind to the class I HLA molecules: HLA-A2 and HLA-A*0101 and B*0801 and HLA-A2+ CD patients have an increased number of specific gluten-specific, CD8+ cytotoxic T lymphocytes (CTLs) in lamina propria which express IFNγ, CD95 and granzyme B (GZMB) when stimulated with gliadin [12,13,14]. However, it should be noted that there is little direct association between CD and specific HLA class I alleles [15], suggesting that antigen-specific CTL activity is not part of the genetic susceptibility to CD.
A hallmark of active CD is an increased number of IELs in the small intestine, even when pathology is not severe [16]. These IELs comprise cytotoxic CD8+ T lymphocytes (CTL), γ/δ T cells and NK cells. Similar cells are found in the lamina propria of the small intestine of CD patients [11,13,17] and the mucosa of untreated CD contains high levels of cytokines that can potentiate cytotoxic lymphocyte activity such as IL-15, IL-21, type I and II Interferons (IFNs) [18,19,20,21].
IELs constitute the largest T cell compartment in the body with one IEL being found for every 10 epithelial cells in the human small intestine [16,22]. Two main populations of IEL are present, adaptive (or conventional) and innate-like or unconventional IELs [23]. Adaptive IELs are TCRαβ+ T cells with a memory phenotype and consist of CD8αβ+ (~80%) and CD4+ (~10%) subsets, all of which are derived from naïve T cells that have been primed by specific peptide antigens presented by MHC on DC in secondary lymphoid organs. In contrast, innate-like IELs are activated by cytokines (IL-15) and NK receptors, in a TCR-independent manner, and can represent 5–30% of human IELs [22]. Innate-like IELs include both TCRαβ+ CD8αα+ and TCRγ/δ+ T cells. A hallmark of untreated CD is an expanded population of TCRγ/δ+ IELs that recognise the non-classical class I MHC molecules, butyrophilin-like (BTNL) molecules BTNL3/8, and whose numbers remain high even after long period on a gluten-free diet (GFD) [24].
As with NK cells, innate-like IELs can be activated in CD via recognition of non-classical class I MHC molecules such as MIC-A on stressed epithelial cells by the NK cell receptor NKG2D on IELs [24,25,26,27,28]. NKG2C/CD94 is a further NK cell receptor, which interacts with the non-classical class I MHC molecule HLA-E present on epithelial cells exposed to IFNs and other inflammatory cytokines [29,30]. The NKG2C/CD94-HLA-E interaction is thought to induce IFNγ production by CTLs and enhance their cytotoxic activity in CD patients [17,31,32]. Type I IFNs may also potentiate the non-antigen-specific cytotoxic activity of CTLs against epithelial cells [21].

1.2.3. Innate Immunity

In addition to antigen-specific CD4+ T cell mediated immunity, several other aspects of the immune response are involved in CD. Recent studies have underlined the critical importance of external factors triggering innate immunity in the pathogenesis of CD. These include viral infections [9], dysbiosis of the microbiota [33], amylase-trypsin inhibitors [34] and a non-T cell epitope peptide of gliadin (known as p31-43) [35]. Innate immunity and chronic inflammation have a direct impact on potentiating the activation of both gluten-specific CD4+ T cell and IELs in CD [9,24,36].
Several proinflammatory effects have been described for p31-43 peptide in vitro and in vivo [35]. It does not bind to the HLA-DQ2 or DQ8 molecules [37], and a specific surface receptor has not been identified [38]. However, it has been shown that p31-43, binds to the nucleotide-binding domain 1 (NBD1) subunit of the cystic fibrosis transmembrane conductance regulator (CFTR), reducing its ATPase activity and causing an increase in reactive oxygen species (ROS) generation and a persistent activation of TG2, leading to increased nuclear translocation of NFκB. Activation of the NFκB pathway induces the transcription of pro-inflammatory cytokines such as IL-17A, IL-21 and IL-15, together with production of active IL-1β [39]. Some of these effects may reflect the fact that p31-43 has a poly-proline II structure and forms oligomers stable in solution with pH ranging from 4 to 8 [40,41], resulting in endoplasmic reticulum-stress (ER-stress), the production of ROS, release of proinflammatory mediators such as IL-1β, and the induction of cell death [35,42].

2. Apoptosis and Cell Shedding from the Epithelium

Under homeostatic conditions, the epithelial layer of the human small intestine is almost completely renewed every week, with a balance between extrusion of effete enterocytes at the tip of the villi and the production of new cells in the transit-amplifying zone of the crypts [43]. As in other epithelial tissues, the loss of enterocytes from the crowded villus tip is triggered by the stretch-activated channel protein Piezo 1 [44] which leads to the release of sphingosine-1-phosphate (S1P) that drives the release of the cells from integrin-dependent anchoring to the extracellular matrix. This leads to a form of apoptotic cell death known as anoikis and extrusion of cells from the epithelium [44,45]. Inflammatory mediators such as TNFα and IFNγ, as well as microbial associated molecular patterns (MAMPs) and pathogens may increase the speed of this process.
Although this mechanism has not been assessed in CD patients, the presence of high concentrations of IFNγ in celiac mucosa is likely to result in the loosening of the tight junctions between epithelial cells and defective closing over of the extrusion area left by shedding of dying cells. In support of this idea, the junctional protein E-cadherin is essential for closing these exposed spots by elongating neighboring cells, which avoids the formation of transient epithelial gaps [46,47] and its expression is downregulated in epithelial cells exposed to IFNγ and TNFα in vitro [48], as well as in CD [49] and IBD in vivo [44,50].
Apoptosis can occur by intrinsic and extrinsic pathways which trigger a common executioner mechanism (summarized in Figure 1).

2.1. Intrinsic Apoptosis

The intrinsic pathway involves permeabilization of the mitochondria outer membrane (MOMP) due to activation of the pore forming proteins BAK (BCL2 antagonist/killer 1) and BAX (BCL2 associated X, apoptosis regulator). This pore facilitates the release of cytochrome-c that binds and activates apoptotic peptidase activating factor (APAF-1) and the initiator caspase-9, together forming the apoptosome which promotes activation of the executioner caspase-3. Caspase-3 activates other procaspases (e.g., caspase-2, -6, -8, and -10), creating an apoptosis-amplifying cascade, which ends in the alteration of the nuclear membrane, the cleavage of intracellular proteins (e.g., PARP), membrane blebbing, and the breakdown of genomic DNA into nucleosomal structures. Release of SMAC (second mitochondria-derived activator of caspases) and the HtrA serine protease 2 (HTRA2) from the mitochondrial membrane also leads to inhibition of the caspase inhibitor X-linked inhibitor of apoptosis protein (XIAP), thus enhancing and sustaining activation of caspase-3. The mitochondrial events in apoptosis are regulated by a balance between pro- and anti-apoptotic proteins of the BCL family. The “BH3-only” domain, t-BID, BIM, NOXA, and PUMA are pro-apoptotic, as they interact with BAK and BAX and promote their membrane permeabilizing actions. On the other hand, proteins belonging to BCL-2 family (BCL-2, BCL-XL, BCL-W, BFL1, MCL1) inhibit apoptosis by competing with the binding of the BH3-only family members to BAX and BAK [51,52].
The intrinsic pathway is triggered by factors such as damage to genomic or mitochondrial DNA, signals derived from damaged organelles (e.g., ER or mitochondrial stress), the inhibition or reduction of growth factors, and the inhibition of intracellular signaling pathways [51]. One of the core mediators of intrinsic apoptosis is the p53 protein, which is activated upon DNA damage induced by oxidants, alkylating agents, or radiation, and it promotes the expression of the pro-apoptotic factors BAX, PUMA and NOXA, while downregulating the expression of anti-apoptotic BCL-2.
One of the most potent immunological triggers for intrinsic apoptosis is the lethal hit delivered by cytotoxic T lymphocytes and NK cells. These cells release granules containing GZMB and Perforin-1 (PRF1), with PRF1 inducing uptake of GZMB via membrane pores and GZMB, inducing apoptosis by cleaving and activating caspases-3, -7, -8 and -10 and BID [53]. That intrinsic apoptosis may take place in CD is suggested by findings of decreased expression of BCL-2 and increased expression of cytoplasmic tumor protein p53 in the crypts of duodenal tissue from untreated CD patients [54]. Moreover, mRNA levels for BAK (but not BAX) are increased in the duodenum of CD patients, while BAK protein is upregulated in the intestinal epithelium of untreated CD patients and its expression correlates with IFNγ levels [55]. By promoting upregulation of BAK and downregulation of BCL-2, IFNγ has a pro-apoptotic role in epithelial cells in CD [56].
As noted above, an increased number of gliadin-specific, HLA-A2 restricted CTLs has been found in the lamina propria during CD [13], and CTLs of this kind have been shown to cause apoptosis in a model of intestinal epithelial cells in vitro. In addition, an increase in apoptotic enterocytes has been found to correlate with higher cytotoxic activity of IELs in CD and may be associated with higher levels of type I IFNs [21].

2.2. Extrinsic Apoptosis

The extrinsic apoptotic pathway is initiated by receptors containing death domains (DD), specifically the TNF family receptors TNFR1, TNFRS10A/B, death receptor 3 (DR3, known as TLIA), and CD95 (also known as FAS). Signals from outside the cells, TNFα, TRAIL, CD95-Ligand (known as FAS-L), bind to these receptors and trigger the assembly of several intracellular multi-protein complexes which ultimately leads to the activation of caspase-8 and caspase-3. In the case of TNFR1, an initial complex named “Complex I”, comprises the adaptor protein TRADD (TNFRSF1A associated via DD) and several additional proteins (TRAF2, TRAF6, c-IAP1, c-IAP2, RIPK1, LUBAC, SHARPIN, HOIL-1 and HOIP). Ligand binding of FAS, DR3, induces the complex named DISC (death-inducing signal complex), made up by the adaptor protein FAS associated via DD (FADD), caspase-8 or 10 (depending on the cell type) and cFLIP (CASP8 and FADD-like apoptosis regulator). These signaling pathways are regulated by the anti-apoptotic factors such as the long isoform of cFLIP (cFLIP-L) and c-IAPs, the trimerization level of the death receptors, as well as by post translational modification of the protein complexes [57]. Caspase-8 is activated by these pathways via autoproteolysis, leading to the generation of cleaved caspase-8 (CC8), which then activates caspase-3, either directly, or indirectly by cleaving BID and promoting MOMP [57].
Activated CTL and NK cells can trigger the extrinsic pathway of apoptosis via the expression of CD95L and TRAIL [58,59,60]. Furthermore, increased expression of CD95 and CD95L has been found in epithelial cells of untreated CD patients [61] and this can be upregulated further by treatment of duodenal biopsies from CD patients with gliadin peptides [62]. In addition, type I and II IFNs may play a role in sensitizing epithelial cells to the extrinsic apoptotic pathway by inducing the expression of both the death receptors and their ligands [63,64].

2.3. Common Executioner Pathway of Apoptosis

Key molecules in the apoptosis process can be detected in the small intestine of untreated CD patients, as well as in a mouse model of gliadin-driven innate immunity. Activation of capase-3 is a key event in the common pathway of apoptosis [57] and increased levels of cleaved caspase-3 (CC3) have been found in epithelial cells from intestinal biopsies of untreated CD patients [54]. Recently, we have confirmed these findings and have found that levels of activated caspases-8 and -3 are also increased in newly diagnosed CD patients [65]. In addition, M30, which marks a CC3-degraded form of cytokeratin-18, correlates with CC3+ epithelial cells and both M30 and CC3 co-stain with Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)+ apoptotic epithelial cells at the villus tip in CD [55,61,66]. Increased numbers of M30+ epithelial cells have also been observed after incubating duodenal biopsies of CD patients with type I IFNs [21].
Studies by our group have shown that oral administration of the innate active p31-43 peptide of gliadin induces upregulation of IFNγ, and increases cell death in the small intestine of wild-type mice, paralleled by an increase in the proapoptotic Bax:Bcl-2 ratio at mRNA level [67]. In parallel, administration of p31-43 leads to increased numbers of TUNEL+ cells and increased expression of CC3 by both epithelial and lamina propria cells [42].

3. Non-Apoptotic Forms of Programmed Cell Death and Their Implications for CD

Other programed cell death mechanisms have been described in recent years which have necrosis-like phenotypes and lead to the release of inflammatory mediators [1] such as IL-1β, IL-18, and alarmins, even before cell death occurs [68,69]. Some of these non-apoptotic PCD pathways are potentially involved in the pathogenesis of CD.

3.1. Pyroptosis

Pyroptosis is a lytic form of programmed cell which leads to rapid clearance of damaged cells during infection. Pyroptotic cells exhibit cell swelling and membrane blebbing, associated with the formation of pores in the cell membrane induced by polymerization of N-terminal fragments of Gasdermin-D (GSDMD). These processes are triggered by a canonical pathway involving the activation of inflammasomes, or by a non-canonical pathway driven by the activation of caspases-4 or -5 (caspase-11 in mouse) [1].
Inflammasomes are a cytosolic multiprotein complex comprising either nod-like receptors (NLRs) (NLR family pyrin domain containing proteins NLRP1, 2, 3, 6, and NLRC4), or non-NLRs (absent in melanoma 2 “AIM2”, and IFNγ-inducible protein 16 “IFI16”) [1,70]. Upon activation, these proteins oligomerize with the apoptosis-associated speck-like protein containing a CARD (ASC), which acts as an adaptor protein that binds and induces autoactivation of caspase-1. When activated, caspase-1 cleaves immature pro-IL-1β and pro-IL-18 into their active forms. In addition, active caspase-1 cleaves GSDMD, producing N-terminal fragments which multimerize and bind to the inner cell membrane, leading to the formation of ~20 nm pores that allow the release of mature IL-1β and IL-18, as well as alarmins such as IL-33, IL-1α and HMGB1 [1,70,71]. GSDMD can also be cleaved by a caspases-4/5 (caspase-11 in mouse)-dependent mechanism in the presence of cytosolic LPS. In this case, caspase-4/5 (or caspase-11) oligomerize and auto-activate, leading to the cleavage of IL-1β, IL-18 and GSDMD [1,72]. There is a balance between pore formation and cell membrane repair, and if the number of pores exceeds the capacity of repair, the cell will die by the process of pyroptosis [1].
Activation of the inflammasome requires two signals. First, a “priming signal” such as TLR ligands, IFNs or alarmins are needed to activate the intracellular NF-kB pathway. The second signal can be produced by a broad arrange of stimuli, whose nature depends on the sensor which has been primed (i.e., members of NLR family, AIM2 or IFI16). These can include MAMPs derived from pathogens, particulate materials (silica and asbestos) or DAMPs such as extracellular ATP, uric acid, and cholesterol crystals, or it may involve additional cellular events, including K+ efflux, mitochondrial damage and ROS generation and lysosomal rupture [70,73,74]. Higher levels of ROS [75,76,77], DAMPs such as IL-33 [78] and HMGB1 [79], have been found in increased in CD patients. Interestingly, HMGB1 can promote the activation of pyroptosis by enhancing the delivery of LPS to caspases-4/5 or -11 in the cell cytosol [72].
Components of the local inflammatory response in CD, such as type I and II IFNs can induce the expression of pyroptosis-associated caspase-4 via Interferon Regulatory Factor 1 (IRF1) activation [80], and of the AIM2 inflammasome and IL-18 production via STAT1 [81]. IL-17A, a cytokine upregulated in some CD cases can also induce the expression of NLRP2 and caspase-5 [80].
That these pathways may be participating in CD is suggested by the findings that IL-18 production is increased in small intestinal crypt cells in CD [82] and that there are high levels of circulating IL-18 in untreated CD patients [83]. IL-1β is also found in the supernatants of Peripheral blood mononuclear cells (PBMC) from CD patients upon pepsin-trypsin-treated gliadins exposure [84] and serum levels of IL-1β fall in CD patients after 1 year on a GFD [85].
Similarly, studies using PBMC from CD patients treated with pepsin digested gliadins have shown higher production of IL-1β and IL-18 in cells from CD patients compared with healthy controls. Release of these cytokines was inhibited by blocking K+ efflux, suggesting the role of inflammasome in response to digested gliadins in PBMC from CD patients [86].
In vitro work shows that IFNα stimulates caspase-dependent inflammasome activation and IL-18 production in duodenal biopsies from CD patients in vitro, leading to IL-18 dependent IFNγ production [21]. IFNγ itself has also been shown to induce increased expression of NLRP6, and caspases-1 and -5 in enterocytes isolated from CD patients [80].
As noted above, the p31-43 gliadin peptide induces type I IFN production and local inflammation in the small intestine of wild-type mice when given orally, and we have shown that the resulting pathology is dependent on NLRP3 signaling, leading to activation of caspase-1 and IL-1β [42]. Oligomerization of the p31-43 peptide may be responsible for providing signal 2 for inflammasome activation in this context [40].
Interestingly, a group of CD patients share an SNP (rs 12150220 A/A) located in the coding region of NLRP1. This SNP is also found in other inflammatory conditions and is associated with increased IL-18 levels in serum, perhaps accounting for the increased levels of IL-18 seen in some CD patients [87]. A SNP in the IL-1β gene (rs16944 C > T, also known as IL1B -511T) has also been found to be associated with osteopenia/osteoporosis and lower mineral density in CD patients [88]. This SNP is thought to influence the expression levels of IL-1β and is also associated with higher risk of other chronic inflammatory conditions such as Alzheimer’s disease [89], gastric cancer [90], keratoconus [91] and Grave’s disease [92].
Thus, there is evidence in both humans and mice that inflammasome activation and pyroptosis may occur in the small intestine in CD (summarized in Figure 2). As well as potentially explaining some of the epithelial cell death, this mechanism may contribute to other aspects of CD pathogenesis, including the increased production and release of IL-18 and IL-1β, both of which can produce inflammation directly and can activate other immune cells, including Th1, Th17 and CTLs [93,94,95]. IL-1β can also induce expression of a FOXP3 splice variant (FOXP3Δ2Δ7) which is associated with poor regulatory T cell function [96,97] and is associated with CD [98]. The alarmins HMGB1 [79], IL-1α [85] and IL-33 [78,99] are also released during pyroptosis and have been associated with CD, where they may play a number of roles (see below).

3.2. Necroptosis

Necroptosis is a necrotic PCD mechanism, which involves the phosphorylation and activation of the membrane pore protein mixed lineage kinase domain-like pseudokinase (MLKL) by activated receptor interacting serine/threonine kinase 3 (RIPK3) [1]. RIPK3 phosphorylation and subsequent activation can be triggered by a broad range of stimuli, including the death receptor ligands that trigger extrinsic apoptosis (TNFα, CD95L, TRAIL). Their ability to induce necroptosis is dependent on apoptosis being blocked, for instance by the presence of caspase-8 inhibitors, or when its activation is prevented by inhibition of the adaptor protein FADD [57,100,101]. When enzymatic activity of caspase-8 is deficient, phospho-RIPK1 binds RIPK3, producing an intracellular protein complex (the necrosome) that recruits and activates MLKL. This leads to the vesicular transport of phospho-MLKL (pMLKL) to the plasma membrane, where it binds to the inner cellular membrane and forms a pore [102]. When MLKL activation exceeds the membrane repair capacity, the loss of membrane integrity triggers necrosis and the release of alarmins such as ATP, HMGB1, IL-1α and IL-33. RIPK1-independent necroptosis can also occur when RIPK3 is activated by viral dsRNA-mediated activation of the TLR3-TRIM pathway [103] and by activation of Z-DNA Binding Protein 1 (ZBP1) [104,105].
Untreated CD patients showed significantly higher mRNA expression of RIPK3, ZBP1 and MLKL, suggesting that necroptosis may account in part for the increased cell death found in active CD [65]. Furthermore, oral administration of gliadin p31-43 led to increased cell death and RIPK3 expression in small intestinal epithelial and lamina propria cells in wild-type mice [42]. Necroptosis was also observed in Paneth cells in inflamed ileal tissues from patients with Crohn’s disease, and experiments in mice demonstrated that Paneth cells follow a necroptosis pathway, via IFNs/STAT1 and MLKL, controlled by caspase-8 [106].
After necrosome formation, pMLKL is transported inside vesicles to the plasma membrane escorted by proteins involved in the structure of tight junctions, and among them is Zonula occludens-1 (ZO-1), which can inhibit pore formation by pMLKL [102]. Deposition of MLKL pores and necroptosis in enterocytes due to downregulation of ZO-1 in untreated CD is thought to contribute to the loosening of tight junctions and loss of the integrity of the epithelial barrier [107,108].
Although some of the pro-necroptotic factors and positive modulators of necroptosis have been found in CD (summarized in Figure 3, Top Image), further investigation is needed to determine the role of necroptosis in this pathology.

3.3. Ferroptosis

Ferroptosis is a non-apoptotic-like PCD, with exclusive biochemical and morphological changes [109] where iron ions catalyze oxidative reactions on poly-unsaturated fatty acids (PUFA) mainly found in the mitochondrial and plasma membrane. The phenotype of these cells differs from classical necrotic or apoptotic cells with a characteristic dysmorphic permeabilized outer membrane of mitochondria and damaged plasma membrane occurring due to excessive oxidation of membrane lipids [1,110,111,112]. This new form of PCD leads to the release of DAMPs into the extracellular space (summarized in Figure 3, Bottom Image) [113]. Ferroptosis is induced upon inhibition of the phospholipid peroxidase and oxidoreductase glutathione peroxidase 4 (GPX4) enzyme and reduction of GPX4 substrate, glutathione (GSH) [1,114]. GPX4 is a selenium-enzyme which is part of the major protective mechanism against lipid peroxidation, it uses GSH to reduce H2O2, organic and lipid peroxides. Thus, GPX4 protects cell membranes from the hazards of oxidants molecules, and as a result, the inhibition of GPX4 increases lipid peroxidation. Ferroptosis can also occur secondary to an increase in the intracellular free iron pool (iron overload) which leads to increased H2O2 levels via a series of Fenton reactions [1,69,110].
Increases of ROS, total lipid hydro-peroxides (LOOH) and reduced antioxidant processes have been reported in the peripheral blood and duodenal mucosa of CD patients and these changes are reverted by a GFD [76,77]. In parallel, ROS and nitric oxide production are increased in circulating erythrocytes and the small intestine of untreated CD patients, together with reduced levels of GSH, the principal substrate of GPX4 [75,76,77]. Increases in other markers of oxidation, including catalase, superoxide dismutase, myeloperoxidase and DNA instability have also been detected in peripheral blood of untreated CD patients [115]. A further factor which could lead to reduced GPX4 activity in untreated CD may be the deficiency in selenium uptake which occurs in these patients [116]. Since IFNγ and p53 inhibit the expression of membrane cellular cysteine transporters (SLC3A2 and SLC7A11) [117,118], it may potentiate the induction of ferroptosis in CD.

4. Alarmins and DAMPs

DAMPs (damage-associated molecular patterns) are a set of highly immunogenic molecules which are associated with cell damage, including Type I IFNs, IL-15, which have been linked to CD [17,69,119,120]. Alarmins, a subgroup of DAMPs, are endogenous molecules released upon cell damage (during spontaneous necrosis, necrosis PCD or necrosis-like PCD) that trigger a response on different immune and non-immune cells [2,121]. The release of DAMPs may potentiate and expand the inflammatory process even at distant sites [122] (summarized in Figure 4). Here, we are going to describe HMGB1 and IL-33, which are released under necrotic cell death and have been already associated with CD.

4.1. HMGB1

The high-mobility group box 1 protein (HMGB1) is normally found in the nucleus of a variety of cells where it acts in different DNA repair mechanisms and increases the affinity of several transcription factors to its cognate DNA sequences [123]. However, under certain inflammatory situations, HMGB1 can be relocated in the cytoplasm and then secreted by lysosomal traffic or during necrosis or necrosis-like PCDs [124,125]. Free HMGB1 binds to TLR4 and induces inflammatory reactions depending in the redox state of its cysteine residues [126]. Interestingly, HMGB1 can also bind to LPS and serves as a carrier to trigger TLR4 activation. On the other hand, the receptor for advanced glycation end products (RAGE) is a HMGB1 receptor, which triggers activation of endothelial cell and smooth muscle cell proliferation [127,128]. HMGB1 can also have chemotactic activity, and immune cell activation through TLR4, RAGE and other immune-related receptors [129]. HMGB1 can activate pyroptosis by its capacity to permeabilize endo-lysosomal membranes at low pH and releasing LPS and cathepsin-B into the cytoplasm. This process leads to the activation of several inflammasome sensors by cathepsin-B or caspases-4 or -5 (caspase-11 in mice) by direct binding with LPS (summarized in Figure 2 and Figure 4) [72,130].
Increased levels of HMGB1 have been found in the serum and feces of untreated CD patients [131,132,133], as well as in the blood during autoimmune disorders associated with CD, such as type I diabetes mellitus, Sjogren’s syndrome, and autoimmune thyroiditis [134,135,136,137,138,139]. Moreover, HMGB1 was found to be associated with an increased capacity of dendritic cells to promote a pro-Th1 phenotype in T cells during antigen presentation, to expand CTLs cell populations, and to promote M1 polarization of macrophages (Figure 4) [140,141,142], all this highlights HMGB1’s role in driving inflammation. Thus, these findings suggest that HMGB1 could play a role in the inflammatory response in CD, and this needs further investigation.

4.2. IL-33

IL-33 is a member of the IL-1 family which is normally located in the nucleus of mesenchymal and epithelial cells, but it can be actively released from viable cells or passively from cells undergoing inflammatory PCD; this mechanism is enhanced if IL-33 is translocated to the cytoplasm by unknown mechanisms [143,144,145]. Normally, IL-33 release is prevented during apoptosis by cleavage of IL-33 into inactive fragments by caspases-3 and -7 [146]. By acting via its specific receptor IL-33Rα (also known as ST2L) [147], has a variety of effects on immune cells (summarized in Figure 4) and is known particularly for its role in allergy and parasite infections because of its ability to activate ILC2, Th2 and mast cells [148,149,150]. However, IL-33 can also promote Th1 and importantly, pro-cytotoxic CD8+ T cell activity during viral infections and immune responses to tumors [151,152], and can maintain survival of regulatory T cells [153]. Notably, IL-33 also has an effect on intestinal epithelial cells, inducing their proliferation and stimulating secretion of protective antimicrobial peptides from enterocytes [154,155]. We and others have shown an increase in IL-33 expression in the serum of untreated CD patients [78,99]. Moreover, we have found a large number of different cells associated with microvasculature (characterized by the expression of SMA, CD31 and CD90), with nuclear IL-33 location and others with a cytoplasmic accumulation of this cytokine. Western blot analysis of duodenal mucosa from CD patients also revealed increased levels of the 18–21 kDa sized fragments of IL-33 [78] that are produced by the action of enzymes released by activated neutrophils and mast cells and which have increased affinity for the IL-33R [156,157,158]. In parallel, increased numbers of CD8+IL-33R+ cells are found in duodenal mucosa in untreated CD, suggesting that these free IL-33 fragments could potentiate the cytotoxic actions of CTLs in CD patients [78]. Additionally, IL1RL1, the gene that codes for ST2, has been linked with CD disease SNP (rs1420106) [159]. Moreover, isolated gluten-specific T cells clones overexpressed IL1RL1 after proper gluten challenge [159]. These findings highlight a potential role of IL-33 axis in CD patients.

5. Potential Interplay between Different PCDs

As we have discussed, untreated CD patients have increased numbers of cells dying via apoptosis and by other pro-inflammatory PCD pathways.
Cell death pathways also are interconnected and influence each other. For instance, there is evidence that lipid peroxidation changes associated with ferroptosis may occur in CD [75,76,77,115] and it has been shown that these mediators can induce apoptosis in the presence of a competent antioxidant system (GSH and thioredoxin systems). However, when the antioxidant system is deficient, both apoptosis and pro-inflammatory PCDs may occur because oxidative conditions activate factors such as MLKL, RIPK1/3, NLRP3, caspase-1 and GSDMD. In contrast, the activation of pro-apoptotic caspases-3 and -7 requires effective antioxidant mechanisms [160]. Thus, CD patients that have a deficit in antioxidant capacity [76,77] may develop a greater activation of pro-inflammatory PCDs. On the other hand, CD patients have been shown to have enhanced IFNγ-mediated induction of thioredoxin (Trx1)-dependent antioxidant systems [161], and Trx1 is thought to support the activation of TG2 activity [162]. Further studies on the oxidative mechanism are needed to assess its relevance in modulating PCDs in CD patients.
The co-existence of multiple cell death pathways has been found in other inflammatory conditions such as Crohn’s disease [106], and in an enteropathy experimental model induced by a single dose of p31-43 [42]. This phenomenon has been referred to as PANoptosis and it is defined as the outcome of evolutionary conserved, interrelated pathways of cell death, which leads to different outcomes (apoptosis, necroptosis and pyroptosis) [163]. Furthermore, it is suggested that this process involves a unique pathway controlled by a multiprotein complex called the “PANoptosome” based on ZBP1. Inflammatory responses driven by activation of the PANoptosome have been postulated in neurodegenerative diseases, cancer, infection-driven inflammation, joint inflammation, and metabolic inflammation [163].
In turn, cytokines and alarmins may modulate PCDs pathways. IFNs are potent modulators of different PCDs, by stimulating the expression of proapoptotic proteins such as BAK, NOXA, caspase-8, and death receptors or death ligands (TNFα, CD95L), while promoting also anti-apoptotic proteins such as cFLIP [64,164]. IFNs may also control the expression of pyroptotic proteins such as caspases-4 and -5, caspase-1 and AIM2, NLRP6, NLRC5 [80,81]. Additionally, IFNs trigger necroptosis by the STAT1 dependent pathway or by increasing ZBP1 and MLKL [104,106,165]. As discussed above, the inflammatory processes triggered in CD may enhance cell death mechanisms of different kinds, leading to the release of HMGB1, IL-1β and IL-33, which expand the inflammation and the induction of further proinflammatory cell death (summarized in Figure 4). Nutritional deficiencies due to mucosal malabsorption and inflammatory process are associated with a broad range of extraintestinal conditions (dermatological, endocrine, and reproductive disorders, neurological and psychiatric conditions, musculoskeletal manifestations, among others) in CD patients [166]. Therefore, inflammation beyond the small intestine deserves further investigation, in order to gain insight into the mechanisms playing a role in proximal small intestines from CD patients, which may be involved in driving systemic disorders.

6. Conclusions

The past few years have seen an expansion in the knowledge about programmed cell death, not only in a detailed description of the molecular mechanisms, but also in the discovery of new pathways occurring under specific conditions. This broad field has an immense impact in health and disease. Particularly, different programmed cell death pathways may occur in enterocytes and lamina propria cells from the small intestine. In addition to silent apoptosis driven by cytotoxic lymphocytes, proinflammatory processes such as pyroptosis, ferroptosis and necroptosis can also be induced. The alarmins and other pro-inflammatory mediators released by these PCD pathways may play a role in expanding the local inflammatory reaction and, by sustaining the T cell-driven tissue damage, help to the loss of tolerance to gluten-derived peptides in the small intestine of CD patients. Release of inflammatory mediators by proinflammatory PCD may have also a role in potentiating and expanding the tissue damage locally and at distant sites, which may favor the development of chronic inflammatory processes and autoimmunity in susceptible individuals. Control of these PCD pathways may have a therapeutic benefit. However, further efforts will need to validate findings from animal models to human diseases.

Author Contributions

F.P.: Conceptualization, co-writing of the original draft and co-design of the Figures. C.N.R.: Conceptualization and editing of the manuscript, co-design of the Figures. E.M. and P.C.: Conceptualization and editing of the manuscript. F.G.C.: Conceptualization, co-writing, review and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the grant PICT 2017 0880 from the Agencia Nacional de Promoción Científica y Tecnológica from Ministerio de Ciencia, Tecnología e Innovación Productiva, República Argentina. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Acknowledgments

We thank to Allan McI. Mowat for critical reading and valuable suggestions of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tang, D.; Kang, R.; Berghe, T.V.; Vandenabeele, P.; Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 2019, 29, 347–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Rider, P.; Voronov, E.D.; Charles, A.; Ron, N. Alarmins: Feel the Stress. J. Immunol. 2017, 198, 1395–1402. [Google Scholar] [CrossRef] [Green Version]
  3. Patankar, J.V.; Becker, C. Cell death in the gut epithelium and implications for chronic inflammation. Nat. Rev. Gastroenterol. Hepatol. 2020. [Google Scholar] [CrossRef]
  4. Evavold, C.L.; Ruan, J.; Tan, Y.; Xia, S.; Wu, H.; Kagan, J.C. The Pore-Forming Protein Gasdermin D Regulates Interleukin-1 Secretion from Living Macrophages. Immunity 2018, 48, 35–44.e6. [Google Scholar] [CrossRef] [Green Version]
  5. Abadie, V.; Sollid, L.M.; Barreiro, L.B.; Jabri, B. Integration of genetic and immunological insights into a model of celiac disease pathogenesis. Annu. Rev. Immunol. 2011, 29, 493–525. [Google Scholar] [CrossRef] [Green Version]
  6. Sollid, L.M.; Lundin, K.E.A. Celiac disease. Autoimmune Dis. 2019, 849–869. [Google Scholar] [CrossRef]
  7. Lindfors, K.; Ciacci, C.; Kurppa, K.; Lundin, K.E.A.; Makharia, G.K.; Mearin, M.L.; Murray, J.A.; Verdu, E.F.; Kaukinen, K. Coeliac disease. Nat. Rev. Dis. Primers 2019, 5, 1–18. [Google Scholar] [CrossRef]
  8. Heyman, M.; Menard, S. Pathways of gliadin transport in celiac disease. Ann. N. Y. Acad. Sci. 2009, 1165, 274–278. [Google Scholar] [CrossRef] [PubMed]
  9. Bouziat, R.; Hinterleitner, R.; Brown, J.J.; Stencel-Baerenwald, J.E.; Ikizler, M.; Mayassi, T.; Meisel, M.; Kim, S.M.; Discepolo, V.; Pruijssers, A.J.; et al. Reovirus infection triggers inflammatory responses to dietary antigens and development of celiac disease. Science 2017, 356, 44–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Mazzarella, G. Effector and suppressor T cells in celiac disease. World J. Gastroenterol. 2015, 21, 7349–7356. [Google Scholar] [CrossRef] [PubMed]
  11. Jabri, B.; Sollid, L.M. T Cells in Celiac Disease. J. Immunol. 2017, 198, 3005–3014. [Google Scholar] [CrossRef] [PubMed]
  12. Gianfrani, C.; Troncone, R.; Mugione, P.; Cosentini, E.; de Pascale, M.; Faruolo, C.; Senger, S.; Terrazzano, G.; Southwood, S.; Auricchio, S.; et al. Celiac Disease Association with CD8+ T Cell Responses: Identification of a Novel Gliadin-Derived HLA-A2-Restricted Epitope. J. Immunol. 2003, 170, 2719–2726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Mazzarella, G.; Stefanile, R.; Camarca, A.; Giliberti, P.; Cosentini, E.; Marano, C.; Iaquinto, G.; Giardullo, N.; Auricchio, S.; Sette, A.; et al. Gliadin Activates HLA Class I-Restricted CD8+ T Cells in Celiac Disease Intestinal Mucosa and Induces the Enterocyte Apoptosis. Gastroenterology 2008, 134, 1017–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Picascia, S.; Sidney, J.; Camarca, A.; Mazzarella, G.; Giardullo, N.; Greco, L.; Auricchio, R.; Auricchio, S.; Troncone, R.; Sette, A.; et al. Gliadin-Specific CD8+ T Cell Responses Restricted by HLA Class I A*0101 and B*0801 Molecules in Celiac Disease Patients. J. Immunol. 2017, 198, 1838–1845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Meresse, B.; Curran, S.A.; Ciszewski, C.; Orbelyan, G.; Setty, M.; Bhagat, G.; Jabri, B. Reprogramming of CTLs into Natural Killer-like Cells in Celiac Disease. J. Exp. Med. 2006, 203, 1343–1345. [Google Scholar] [CrossRef] [PubMed]
  16. Ferguson, A.; Mcclure, J.P.; Townley, R.R.W. Intraepithelial Lymphocyte Counts in Small Intestinal Biopsies From Children With Diarrhoea. Acta Pædiatrica 1976, 65, 541–546. [Google Scholar] [CrossRef]
  17. Meresse, B.; Chen, Z.; Ciszewski, C.; Tretiakova, M.; Bhagat, G.; Krausz, T.N.; Raulet, D.H.; Lanier, L.L.; Groh, V.; Spies, T.; et al. Coordinated induction by IL15 of a TCR-independent NKG2D signaling pathway converts CTL into lymphokine-activated killer cells in celiac disease. Immunity 2004, 21, 357–366. [Google Scholar] [CrossRef] [Green Version]
  18. Abadie, V.; Jabri, B. IL-15: A central regulator of celiac disease immunopathology. Immunol. Rev. 2014, 260, 221–234. [Google Scholar] [CrossRef] [Green Version]
  19. Sarra, M.; Cupi, M.L.; Monteleone, I.; Franzè, E.; Ronchetti, G.; Di Sabatino, A.; Gentileschi, P.; Franceschilli, L.; Sileri, P.; Sica, G.; et al. IL-15 positively regulates IL-21 production in celiac disease mucosa. Mucosal Immunol. 2013, 6, 244–255. [Google Scholar] [CrossRef]
  20. Meresse, B.; Verdier, J.; Cerf-Bensussan, N. The cytokine interleukin 21: A new player in coeliac disease? Gut 2008, 57, 879–881. [Google Scholar] [CrossRef]
  21. Jarry, A.; Malard, F.; Bou-hanna, C.; Meurette, G.; Mohty, M.; Mosnier, J.F.; Laboisse, C.L.; Bossard, C. Interferon-Alpha Promotes Th1 Response and Epithelial Apoptosis via Inflammasome Activation in Human Intestinal Mucosa. Cmgh 2017, 3, 72–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Mayassi, T.; Jabri, B. Human intraepithelial lymphocytes. Mucosal Immunol. 2018, 11, 1281–1289. [Google Scholar] [CrossRef] [PubMed]
  23. Cheroutre, H.; Lambolez, F.; Mucida, D. The light and dark sides of intestinal intraepithelial lymphocytes. Nat. Rev. Immunol. 2011, 11, 445–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mayassi, T.; Ladell, K.; Gudjonson, H.; McLaren, J.E.; Shaw, D.G.; Tran, M.T.; Rokicka, J.J.; Lawrence, I.; Grenier, J.C.; van Unen, V.; et al. Chronic Inflammation Permanently Reshapes Tissue-Resident Immunity in Celiac Disease. Cell 2019, 176, 967–981.e19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Abadie, V.; Discepolo, V.; Jabri, B. Intraepithelial lymphocytes in celiac disease immunopathology. Semin. Immunopathol. 2012, 34, 551–556. [Google Scholar] [CrossRef]
  26. Hüe, S.; Mention, J.J.; Monteiro, R.C.; Zhang, S.L.; Cellier, C.; Schmitz, J.; Verkarre, V.; Fodil, N.; Bahram, S.; Cerf-Bensussan, N.; et al. A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity 2004, 21, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Allegretti, Y.L.; Bondar, C.; Guzman, L.; Cueto Rua, E.; Chopita, N.; Fuertes, M.; Zwirner, N.W.; Chirdo, F.G. Broad MICA/B Expression in the Small Bowel Mucosa: A Link between Cellular Stress and Celiac Disease. PLoS ONE 2013, 8, e73658. [Google Scholar] [CrossRef] [Green Version]
  28. Jabri, B.; De Serre, N.P.M.; Cellier, C.; Evans, K.; Gache, C.; Carvalho, C.; Mougenot, J.F.; Allez, M.; Jian, R.; Desreumaux, P.; et al. Selective expansion of intraepithelial lymphocytes expressing the HLA-E- specific natural killer receptor CD94 in celiac disease. Gastroenterology 2000, 118, 867–879. [Google Scholar] [CrossRef]
  29. Gustafson, K.S.; Ginder, G.D. Interferon-γ induction of the human leukocyte antigen-E gene is mediated through binding of a complex containing STAT1α to a distinct interferon-γ- responsive element. J. Biol. Chem. 1996, 271, 20035–20046. [Google Scholar] [CrossRef] [Green Version]
  30. Gobin, S.J.P.; van den Elsen, P.J. Transcriptional regulation of the MHC class Ib genes HLA-E, HLA-F and HLA-G. Hum. Immunol. 2000, 61, 1102–1107. [Google Scholar] [CrossRef]
  31. Meresse, B.; Meresse, B.; Ciszewski, C.; Setty, M.; Curran, S.; Tretiakova, M.; Krausz, T.; Lanier, L.; Ebert, E.; Green, P.H.; et al. Selective expression of NKG2C in intraepithelial lymphocytes(IEL): A basis for IEL proliferation and epithelial cell killing in celiac disease (CD). J. Pediatr. Gastroenterol. Nutr. 2005, 41, 495. [Google Scholar] [CrossRef]
  32. Gumá, M.; Busch, L.K.; Salazar-Fontana, L.I.; Bellosillo, B.; Morte, C.; García, P.; López-Botet, M. The CD94/NKG2C killer lectin-like receptor constitutes an alternative activation pathway for a subset of CD8+ T cells. Eur. J. Immunol. 2005, 35, 2071–2080. [Google Scholar] [CrossRef] [PubMed]
  33. Caminero, A.; Verdu, E.F. Celiac disease: Should we care about microbes? Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G161–G170. [Google Scholar] [CrossRef] [PubMed]
  34. Junker, Y.; Zeissig, S.; Kim, S.J.; Barisani, D.; Wieser, H.; Leffler, D.A.; Zevallos, V.; Libermann, T.A.; Dillon, S.; Freitag, T.L.; et al. Wheat amylase trypsin inhibitors drive intestinal inflammation via activation of toll-like receptor 4. J. Exp. Med. 2012, 209, 2395–2408. [Google Scholar] [CrossRef] [PubMed]
  35. Chirdo, F.G.; Auricchio, S.; Troncone, R.; Barone, M.V. The Gliadin p31-43 Peptide: Inducer of Multiple Proinflammatory Effects, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2021; Volume 358. [Google Scholar]
  36. Abadie, V.; Kim, S.M.; Lejeune, T.; Palanski, B.A.; Ernest, J.D.; Tastet, O.; Voisine, J.; Discepolo, V.; Marietta, E.V.; Hawash, M.B.F.; et al. IL-15, gluten and HLA-DQ8 drive tissue destruction in coeliac disease. Nature 2020, 578, 600–604. [Google Scholar] [CrossRef]
  37. Falcigno, L.; Calvanese, L.; Conte, M.; Nanayakkara, M.; Barone, M.V.; D’auria, G. Structural perspective of gliadin peptides active in celiac disease. Int. J. Mol. Sci. 2020, 21, 9301. [Google Scholar] [CrossRef]
  38. Paolella, G.; Lepretti, M.; Martucciello, S.; Nanayakkara, M.; Auricchio, S.; Esposito, C.; Barone, M.V.; Caputo, I. The toxic alpha-gliadin peptide 31-43 enters cells without a surface membrane receptor. Cell Biol. Int. 2018, 42, 112–120. [Google Scholar] [CrossRef]
  39. Maiuri, L.; Villella, V.R.; Raia, V.; Kroemer, G. The gliadin-CFTR connection: New perspectives for the treatment of celiac disease. Ital. J. Pediatr. 2019, 45, 1–4. [Google Scholar] [CrossRef]
  40. Gómez Castro, M.F.; Miculán, E.; Herrera, M.G.; Ruera, C.; Perez, F.; Prieto, E.D.; Barrera, E.; Pantano, S.; Carasi, P.; Chirdo, F.G. p31-43 Gliadin Peptide Forms Oligomers and Induces NLRP3 Inflammasome/Caspase 1- Dependent Mucosal Damage in Small Intestine. Front. Immunol. 2019, 10, 31. [Google Scholar] [CrossRef] [Green Version]
  41. Herrera, M.G.; Gómez Castro, M.F.; Prieto, E.; Barrera, E.; Dodero, V.I.; Pantano, S.; Chirdo, F. Structural conformation and self-assembly process of p31-43 gliadin peptide in aqueous solution. Implications for celiac disease. FEBS J. 2020, 287, 2134–2149. [Google Scholar] [CrossRef]
  42. Ruera, C.N.; Miculán, E.; Pérez, F.; Ducca, G.; Carasi, P.; Chirdo, F.G. Sterile inflammation drives multiple programmed cell death pathways in the gut. J. Leukoc. Biol. 2021, 109, 211–221. [Google Scholar] [CrossRef]
  43. Williams, J.M.; Duckworth, C.A.; Burkitt, M.D.; Watson, A.J.M.; Campbell, B.J.; Pritchard, D.M. Epithelial Cell Shedding and Barrier Function: A Matter of Life and Death at the Small Intestinal Villus Tip. Vet. Pathol. 2015, 52, 445–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Gudipaty, S.A.; Rosenblatt, J. Epithelial cell extrusion: Pathways and pathologies. Semin. Cell Dev. Biol. 2017, 67, 132–140. [Google Scholar] [CrossRef]
  45. Eisenhoffer, G.T.; Loftus, P.D.; Yoshigi, M.; Otsuna, H.; Chien, C.; Morcos, P.A.; Rosenblatt, J. Crowding induces live cell extrusion to maintain homeostatic cell numbers in epithelia. Nature 2012, 484, 546–549. [Google Scholar] [CrossRef]
  46. Lubkov, V.; Bar-Sagi, D. E-cadherin-mediated cell coupling is required for apoptotic cell extrusion. Curr. Biol. 2014, 24, 868–874. [Google Scholar] [CrossRef] [Green Version]
  47. Kiesslich, R.; Goetz, M.; Angus, E.M.; Hu, Q.; Guan, Y.; Potten, C.; Allen, T.; Neurath, M.F.; Shroyer, N.F.; Montrose, M.H.; et al. Identification of Epithelial Gaps in Human Small and Large Intestine by Confocal Endomicroscopy. Gastroenterology 2007, 133, 1769–1778. [Google Scholar] [CrossRef]
  48. Bruewer, M.; Luegering, A.; Kucharzik, T.; Parkos, C.A.; Madara, J.L.; Hopkins, A.M.; Nusrat, A. Proinflammatory Cytokines Disrupt Epithelial Barrier Function by Apoptosis-Independent Mechanisms. J. Immunol. 2003, 171, 6164–6172. [Google Scholar] [CrossRef] [Green Version]
  49. Ciccocioppo, R.; Finamore, A.; Ara, C.; di Sabatino, A.; Mengheri, E.; Corazza, G.R. Altered expression, localization, and phosphorylation of epithelial junctional proteins in celiac disease. Am. J. Clin. Pathol. 2006, 125, 502–511. [Google Scholar] [CrossRef] [PubMed]
  50. Karayiannakis, A.J.; Syrigos, K.N.; Efstathiou, J.; Valizadeh, A.; Noda, M.; Playford, R.J.; Kmiot, W.; Pignatelli, M. Expression of catenins and E-cadherin during epithelial restitution in inflammatory bowel disease. J. Pathol. 1998, 185, 413–418. [Google Scholar] [CrossRef]
  51. Singh, R.; Letai, A.; Sarosiek, K. Regulation of apoptosis in health and disease: The balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 2019, 20, 175–193. [Google Scholar] [CrossRef] [PubMed]
  52. Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
  53. Afonina, I.S.; Cullen, S.P.; Martin, S.J. Cytotoxic and non-cytotoxic roles of the CTL/NK protease granzyme B. Immunol. Rev. 2020, 235, 105–116. [Google Scholar] [CrossRef] [PubMed]
  54. Shalimar, D.M.; Das, P.; Sreenivas, V.; Gupta, S.D.; Panda, S.K.; Makharia, G.K. Mechanism of villous atrophy in celiac disease: Role of apoptosis and epithelial regeneration. Arch. Pathol. Lab. Med. 2013, 137, 1262–1269. [Google Scholar] [CrossRef] [PubMed]
  55. Cheravsky, A.C.; Rubio, A.E.; Vanzulli, S.; Rubinstein, N.; de Rosa, S.; Fainboim, L. Evidences of the involvement of Bak, a member of the Bcl-2 family of proteins, in active coeliac disease. Autoimmunity 2002, 35, 29–37. [Google Scholar] [CrossRef] [PubMed]
  56. Zhou, Y.; Weyman, C.M.; Liu, H.; Almassan, A.; Zhou, A. IFN-γ induces apoptosis in HL-60 cells through decreased Bcl-2 and increased Bak expression. J. Interf. Cytokine Res. 2008, 28, 65–72. [Google Scholar] [CrossRef]
  57. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
  58. Mirandola, P.; Ponti, C.; Gobbi, G.; Sponzilli, I.; Vaccarezza, M.; Cocco, L.; Zauli, G.; Secchiero, P.; Manzoli, F.A.; Vitale, M. Activated human NK and CD8+ T cells express both TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors but are resistant to TRAIL-mediated cytotoxicity. Blood 2004, 104, 2418–2424. [Google Scholar] [CrossRef]
  59. Prager, I.; Liesche, C.; Van Ooijen, H.; Urlaub, D.; Verron, Q.; Sandström, N.; Fasbender, F.; Claus, M.; Eils, R.; Beaudouin, J.; et al. NK cells switch from granzyme B to death receptor-mediated cytotoxicity during serial killing. J. Exp. Med. 2019, 216, 2113–2127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Brincks, E.L.; Katewa, A.; Kucaba, T.A.; Griffith, T.S.; Legge, K.L. CD8 T Cells Utilize TRAIL to Control Influenza Virus Infection. J. Immunol. 2008, 181, 4918–4925. [Google Scholar] [CrossRef]
  61. Ciccocioppo, R.; Di Sabatino, A.; Parroni, R.; Muzi, P.; D’Alò, S.; Ventura, T.; Pistoia, M.A.; Cifone, M.G.; Corazza, G.R. Increased Enterocyte Apoptosis and Fas-Fas Ligand System in Celiac Disease. Am. J. Clin. Pathol. 2001, 115, 494–503. [Google Scholar] [CrossRef] [Green Version]
  62. Giovannini, C.; Matarrese, P.; Scazzocchio, B.; Varì, R.; D’Archivio, M.; Straface, E.; Masella, R.; Malorni, W.; De Vincenzi, M. Wheat gliadin induces apoptosis of intestinal cells via an autocrine mechanism involving Fas-Fas ligand pathway. FEBS Lett. 2003, 540, 117–124. [Google Scholar] [CrossRef] [Green Version]
  63. Siegmund, D.; Wicovsky, A.; Schmitz, I.; Schulze-Osthoff, K.; Kreuz, S.; Leverkus, M.; Dittrich-Breiholz, O.; Kracht, M.; Wajant, H. Death Receptor-Induced Signaling Pathways Are Differentially Regulated by Gamma Interferon Upstream of Caspase 8 Processing. Mol. Cell. Biol. 2005, 25, 6363–6379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kotredes, K.P.; Gamero, A.M. Interferons as inducers of apoptosis in malignant cells. J. Interferon Cytokine Res. 2013, 33, 162–170. [Google Scholar] [CrossRef] [Green Version]
  65. Ruera, N.C.; Perez, F.; Miculan, E.; Ducca, G.; Guzman, L.; Garbi, L.; Carasi, P.; Chirdo, F. Inflammatory cell death pathways in celiac disease. in preparation.
  66. Moss, S.F.; Attia, L.; Scholes, J.V.; Walters, J.R.F.; Holt, P.R. Increased small intestinal apoptosis in coeliac disease. Gut 1996, 39, 811–817. [Google Scholar] [CrossRef] [Green Version]
  67. Araya, R.E.; Gomez Castro, M.F.; Carasi, P.; McCarville, J.L.; Jury, J.; Mowat, A.M.; Verdu, E.F.; Chirdo, F.G.; Florencia, M.; Castro, G.; et al. Mechanisms of innate immune activation by gluten peptide p31-43 in mice. Am. J. Physiol. Liver Physiol. 2016, 311, G40–G49. [Google Scholar] [CrossRef]
  68. Kuriakose, T.; Kanneganti, T.D. Gasdermin D Flashes an Exit Signal for IL-1. Immunity 2018, 48, 1–3. [Google Scholar] [CrossRef] [Green Version]
  69. Sarhan, M.; Land, W.G.; Tonnus, W.; Hugo, C.P.; Linkermann, A. Origin and consequences of necroinflammation. Physiol. Rev. 2018, 98, 727–780. [Google Scholar] [CrossRef] [PubMed]
  70. Kesavardhana, S.; Kanneganti, T.D. Mechanisms governing inflammasome activation, assembly and pyroptosis induction. Int. Immunol. 2017, 29, 201–210. [Google Scholar] [CrossRef]
  71. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 1–22. [Google Scholar] [CrossRef] [PubMed]
  72. Deng, M.; Tang, Y.; Li, W.; Wang, X.; Zhang, R.; Zhang, X.; Zhao, X.; Liu, J.; Tang, C.; Liu, Z.; et al. The Endotoxin Delivery Protein HMGB1 Mediates Caspase-11-Dependent Lethality in Sepsis. Immunity 2018, 49, 740–753.e7. [Google Scholar] [CrossRef] [Green Version]
  73. Lamkanfi, M.; Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157, 1013–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Li, C.; Chen, M.; He, X.; Ouyang, D. A mini-review on ion fluxes that regulate NLRP3 inflammasome activation. Acta Biochim. Biophys. Sin. 2021, 53, 131–139. [Google Scholar] [CrossRef] [PubMed]
  75. Moretti, S.; Mrakic-Sposta, S.; Roncoroni, L.; Vezzoli, A.; Dellanoce, C.; Monguzzi, E.; Branchi, F.; Ferretti, F.; Lombardo, V.; Doneda, L.; et al. Oxidative stress as a biomarker for monitoring treated celiac disease article. Clin. Transl. Gastroenterol. 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  76. Kasapovi, J.; Peji, S.; Gavrilovi, L.; Radlovi, N.; Saii, Z.; Pajovi, S. Antioxidant status of the celiac mucosa: Implications for disease pathogenesis. In Celiac Disease—From Pathophysiology to Advanced Therapies; InTech: London, UK, 2012. [Google Scholar] [CrossRef] [Green Version]
  77. Stojiljković, V.; Pejić, S.; Kasapović, J.; Gavrilović, L.; Stojiljković, S.; Nikolić, D.; Pajović, S.B. Glutathione redox cycle in small intestinal mucosa and peripheral blood of pediatric celiac disease patients. An. Acad. Bras. Cienc. 2012, 84, 175–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Perez, F.; Ruera, C.N.; Miculan, E.; Carasi, P.; Dubois-Camacho, K.; Garbi, L.; Guzman, L.; Hermoso, M.A.; Chirdo, F.G.; Miculán, E.G.; et al. IL-33 Alarmin and Its Active Proinflammatory Fragments Are Released in Small Intestine in Celiac Disease. Front. Immunol. 2020, 11, 1–15. [Google Scholar] [CrossRef]
  79. Manti, S.; Cuppari, C.; Tardino, L.; Parisi, G.; Spina, M.; Salpietro, C.; Leonardi, S. HMGB1 as a new biomarker of celiac disease in children: A multicenter study. Nutrition 2017, 37, 18–21. [Google Scholar] [CrossRef]
  80. Pietz, G.; De, R.; Hedberg, M.; Sjöberg, V.; Sandström, O.; Hernell, O.; Hammarström, S.; Hammarström, M.-L.L. Immunopathology of childhood celiac disease—Key role of intestinal epithelial cells. PLoS ONE 2017, 12, e0185025. [Google Scholar] [CrossRef] [PubMed]
  81. Cornut, M.; Bourdonnay, E.; Henry, T. Transcriptional regulation of inflammasomes. Int. J. Mol. Sci. 2020, 21, 8087. [Google Scholar] [CrossRef]
  82. León, A.J.; Garrote, J.A.; Blanco-Quirós, A.; Calvo, C.; Fernández-Salazar, L.; Del Villar, A.; Barrera, A.; Arranz, E.; León, A.J.; Garrote, J.A.; et al. Interleukin 18 maintains a long-standing inflammation in coeliac disease patients. Clin. Exp. Immunol. 2006, 146, 479–485. [Google Scholar] [CrossRef] [PubMed]
  83. Merendino, R.A.; Di Pasquale, G.; Sturniolo, G.C.; Ruello, A.; Albanese, V.; Minciullo, P.L.; Di Mauro, S.; Gangemi, S. Relationship between IL-18 and sICAM-1 serum levels in patients affected by coeliac disease: Preliminary considerations. Immunol. Lett. 2003, 85, 257–260. [Google Scholar] [CrossRef]
  84. Harris, K.M.; Fasano, A.; Mann, D.L. Cutting Edge: IL-1 Controls the IL-23 Response Induced by Gliadin, the Etiologic Agent in Celiac Disease. J. Immunol. 2008, 181, 4457–4460. [Google Scholar] [CrossRef] [Green Version]
  85. Manavalan, J.S.; Hernandez, L.; Shah, J.G.; Konikkara, J.; Naiyer, A.J.; Lee, A.R.; Ciaccio, E.; Minaya, M.T.; Green, P.H.R.R.; Bhagat, G. Serum cytokine elevations in celiac disease: Association with disease presentation. Hum. Immunol. 2010, 71, 50–57. [Google Scholar] [CrossRef]
  86. Palová-Jelínková, L.; Dáňová, K.; Drašarová, H.; Dvořák, M.; Funda, D.P.; Fundová, P.; Kotrbová-Kozak, A.; Černá, M.; Kamanová, J.; Martin, S.F.; et al. Pepsin Digest of Wheat Gliadin Fraction Increases Production of IL-1β via TLR4/MyD88/TRIF/MAPK/NF-κB Signaling Pathway and an NLRP3 Inflammasome Activation. PLoS ONE 2013, 8, e62426. [Google Scholar] [CrossRef] [Green Version]
  87. Pontillo, A.; Vendramin, A.; Catamo, E.; Fabris, A.; Crovella, S. The missense variation Q705K in CIAS1/NALP3/NLRP3 gene and an NLRP1 haplotype are associated with celiac disease. Am. J. Gastroenterol. 2011, 106, 539–544. [Google Scholar] [CrossRef] [PubMed]
  88. Moreno, M.L.; Crusius, J.B.A.; Cherñavsky, A.; Sugai, E.; Sambuelli, A.; Vazquez, H.; Mauriño, E.; Peña, A.S.; Bai, J.C. The IL-1 gene family and bone involvement in celiac disease. Immunogenetics 2005, 57, 618–620. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, W.F.; Liao, Y.C.; Wu, S.L.; Tsai, F.J.; Lee, C.C.; Hua, C.S. Association of interleukin-1 beta and receptor antagonist gene polymorphisms with late onset Alzheimer’s disease in Taiwan Chinese. Eur. J. Neurol. 2005, 12, 609–613. [Google Scholar] [CrossRef] [PubMed]
  90. Sultana, Z.; Bankura, B.; Pattanayak, A.K.; Sengupta, D.; Sengupta, M.; Saha, M.L.; Panda, C.K.; Das, M. Association of Interleukin-1 beta and tumor necrosis factor-alpha genetic polymorphisms with gastric cancer in India. Environ. Mol. Mutagen. 2018, 59, 653–667. [Google Scholar] [CrossRef]
  91. Kim, S.H.; Mok, J.W.; Kim, H.S.; Joo, C.K. Association of -31T>C and -511 C>T polymorphisms in the interleukin 1 beta (IL1B) promoter in Korean keratoconus patients. Mol. Vis. 2008, 14, 2109–2116. Available online: http://www.molvis.org/molvis/v14/a247 (accessed on 24 April 2021).
  92. Chen, M.-L.; Liao, N.; Zhao, H.; Huang, J.; Xie, Z.-F. Association between the IL1B (-511), IL1B (+3954), IL1RN (VNTR) Polymorphisms and Graves’ Disease Risk: A Meta-Analysis of 11 Case-Control Studies. PLoS ONE 2014, 9, e86077. [Google Scholar] [CrossRef]
  93. Tsuji-Takayama, K.; Aizawa, Y.; Okamoto, I.; Kojima, H.; Koide, K.; Takeuchi, M.; Ikegami, H.; Ohta, T.; Kurimoto, M. Interleukin-18 induces interferon-γ production through NF-κb and NFAT activation in murine T helper type 1 cells. Cell. Immunol. 1999, 196, 41–50. [Google Scholar] [CrossRef]
  94. Deknuydt, F.; Bioley, G.; Valmori, D.; Ayyoub, M. IL-1β and IL-2 convert human Treg into TH17 cells. Clin. Immunol. 2009, 131, 298–307. [Google Scholar] [CrossRef] [PubMed]
  95. Revu, S.; Wu, J.; Henkel, M.; Rittenhouse, N.; Menk, A.; Delgoffe, G.M.; Poholek, A.C.; McGeachy, M.J. IL-23 and IL-1β Drive Human Th17 Cell Differentiation and Metabolic Reprogramming in Absence of CD28 Costimulation. Cell Rep. 2018, 22, 2642–2653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Mailer, R.K.W. Alternative splicing of FOXP3-virtue and vice. Front. Immunol. 2018, 9, 530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Mailer, R.K.W.; Falk, K.; Rötzschke, O. Absence of leucine zipper in the natural FOXP3Δ2Δ7 isoform does not affect dimerization but abrogates suppressive capacity. PLoS ONE 2009, 4, e6104. [Google Scholar] [CrossRef] [Green Version]
  98. Serena, G.; Yan, S.; Camhi, S.; Patel, S.; Lima, R.S.; Sapone, A.; Leonard, M.M.; Mukherjee, R.; Nath, B.J.; Lammers, K.M.; et al. Proinflammatory cytokine interferon-γ and microbiome-derived metabolites dictate epigenetic switch between forkhead box protein 3 isoforms in coeliac disease. Clin. Exp. Immunol. 2017, 187, 490–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. López-Casado, M.A.; Lorite, P.; Palomeque, T.; Torres, M.I. Potential role of the IL-33/ST2 axis in celiac disease. Cell. Mol. Immunol. 2017, 14, 285–292. [Google Scholar] [CrossRef] [Green Version]
  100. Tsuchiya, Y.; Nakabayashi, O.; Nakano, H. FLIP the switch: Regulation of apoptosis and necroptosis by cFLIP. Int. J. Mol. Sci. 2015, 16, 30321–30341. [Google Scholar] [CrossRef] [Green Version]
  101. Osborn, S.L.; Diehl, G.; Han, S.J.; Xue, L.; Kurd, N.; Hsieh, K.; Cado, D.; Robey, E.A.; Winoto, A. Fas-associated death domain (FADD) is a negative regulator of T-cell receptor-mediated necroptosis. Proc. Natl. Acad. Sci. USA 2010, 107, 13034–13039. [Google Scholar] [CrossRef] [Green Version]
  102. Samson, A.L.; Zhang, Y.; Geoghegan, N.D.; Gavin, X.J.; Davies, K.A.; Mlodzianoski, M.J.; Whitehead, L.W.; Frank, D.; Garnish, S.E.; Fitzgibbon, C.; et al. MLKL trafficking and accumulation at the plasma membrane control the kinetics and threshold for necroptosis. Nat. Commun. 2020, 11, 1–17. [Google Scholar] [CrossRef]
  103. He, S.; Liang, Y.; Shao, F.; Wang, X. Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway. Proc. Natl. Acad. Sci. USA 2011, 108, 20054–20059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yang, D.; Liang, Y.; Zhao, S.; Ding, Y.; Zhuang, Q.; Shi, Q.; Ai, T.; Wu, S.Q.; Han, J. ZBP1 mediates interferon-induced necroptosis. Cell. Mol. Immunol. 2020, 17, 356–368. [Google Scholar] [CrossRef] [PubMed]
  105. Zheng, M.; Kanneganti, T.D. The regulation of the ZBP1-NLRP3 inflammasome and its implications in pyroptosis, apoptosis, and necroptosis (PANoptosis). Immunol. Rev. 2020, 297, 26–38. [Google Scholar] [CrossRef] [PubMed]
  106. Günther, C.; Ruder, B.; Stolzer, I.; Dorner, H.; He, G.W.; Chiriac, M.T.; Aden, K.; Strigli, A.; Bittel, M.; Zeissig, S.; et al. Interferon Lambda Promotes Paneth Cell Death Via STAT1 Signaling in Mice and Is Increased in Inflamed Ileal Tissues of Patients With Crohn’s Disease. Gastroenterology. 2019, 157, 1310–1322.e13. [Google Scholar] [CrossRef] [PubMed]
  107. Pizzuti, D.; Bortolami, M.; Mazzon, E.; Buda, A.; Guariso, G.; D’Odorico, A.; Chiarelli, S.; D’Incà, R.; De Lazzari, F.; Martines, D. Transcriptional downregulation of tight junction protein ZO-1 in active coeliac disease is reversed after a gluten-free diet. Dig. Liver Dis. 2004, 36, 337–341. [Google Scholar] [CrossRef]
  108. Montalto, M.; Cuoco, L.; Ricci, R.; Maggiano, N.; Vecchio, F.M.; Gasbarrini, G. Immunohistochemical Analysis of ZO-1 in the Duodenal Mucosa of Patients with Untreated and Treated Celiac Disease. Digestion 2002, 65, 227–233. [Google Scholar] [CrossRef]
  109. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [Green Version]
  110. Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef] [Green Version]
  111. Riegman, M.; Sagie, L.; Galed, C.; Levin, T.; Steinberg, N.; Dixon, S.J.; Wiesner, U.; Bradbury, M.S.; Niethammer, P.; Zaritsky, A.; et al. Ferroptosis occurs through an osmotic mechanism and propagates independently of cell rupture. Nat. Cell Biol. 2020, 22, 1042–1048. [Google Scholar] [CrossRef]
  112. Yan, B.; Ai, Y.; Sun, Q.; Ma, Y.; Cao, Y.; Wang, J.; Zhang, Z.; Wang, X. Membrane Damage during Ferroptosis Is Caused by Oxidation of Phospholipids Catalyzed by the Oxidoreductases POR and CYB5R1. Mol. Cell 2021, 81, 355–369.e10. [Google Scholar] [CrossRef]
  113. Martin-Sanchez, D.; Ruiz-Andres, O.; Poveda, J.; Carrasco, S.; Cannata-Ortiz, P.; Sanchez-Niño, M.D.; Ruiz Ortega, M.; Egido, J.; Linkermann, A.; Ortiz, A.; et al. Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid-induced AKI. J. Am. Soc. Nephrol. 2017, 28, 218–229. [Google Scholar] [CrossRef] [PubMed]
  114. Xu, Y.; Wang, J.; Song, X.; Qu, L.; Wei, R.; He, F.; Wang, K.; Luo, B. RIP3 induces ischemic neuronal DNA degradation and programmed necrosis in rat via AIF. Sci. Rep. 2016, 6, 1–11. [Google Scholar] [CrossRef]
  115. Maluf, S.W.; Filho, D.W.; Parisotto, E.B.; de Medeiros, G. da S.; Pereira, C.H.J.; Maraslis, F.T.; Schoeller, C.C.D.; da Rosa, J.S.; Fröde, T.S. DNA damage, oxidative stress, and inflammation in children with celiac disease. Genet. Mol. Biol. 2020, 43, 1–8. [Google Scholar] [CrossRef] [PubMed]
  116. Stazi, A.V.; Trinti, B. Selenium status and over-expression of interleukin-15 in celiac disease and autoimmune thyroid diseases. Ann. Ist. Super. Sanita 2010, 46, 389–399. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, W.; Green, M.; Choi, J.E.; Gijón, M.; Kennedy, P.D.; Johnson, J.K.; Liao, P.; Lang, X.; Kryczek, I.; Sell, A.; et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019, 569, 270–274. [Google Scholar] [CrossRef] [PubMed]
  118. Jiang, L.; Kon, N.; Li, T.; Wang, S.J.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef] [Green Version]
  119. Zorro, M.M.; Aguirre-Gamboa, R.; Mayassi, T.; Ciszewski, C.; Barisani, D.; Hu, S.; Weersma, R.K.; Withoff, S.; Li, Y.; Wijmenga, C.; et al. Tissue alarmins and adaptive cytokine induce dynamic and distinct transcriptional responses in tissue-resident intraepithelial cytotoxic T lymphocytes. J. Autoimmun. 2020, 108. [Google Scholar] [CrossRef]
  120. Jabri, B.; Abadie, V. IL-15 functions as a danger signal to regulate tissue-resident T cells and tissue destruction. Nat. Rev. Immunol. 2015, 15, 771–783. [Google Scholar] [CrossRef]
  121. Bianchi, M.E. DAMPs, PAMPs and alarmins: All we need to know about danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef]
  122. Linkermann, A.; Stockwell, B.R.; Krautwald, S.; Anders, H.J. Regulated cell death and inflammation: An auto-amplification loop causes organ failure. Nat. Rev. Immunol. 2014, 14, 759–767. [Google Scholar] [CrossRef]
  123. Yuan, F.; Gu, L.; Guo, S.; Wang, C.; Li, G.M. Evidence for involvement of HMGB1 protein in human DNA mismatch repair. J. Biol. Chem. 2004, 279, 20935–20940. [Google Scholar] [CrossRef] [Green Version]
  124. Lu, B.; Antoine, D.J.; Kwan, K.; Lundbäck, P.; Wähämaa, H.; Schierbeck, H.; Robinson, M.; Van Zoelen, M.A.D.; Yang, H.; Li, J.; et al. JAK/STAT1 signaling promotes HMGB1 hyperacetylation and nuclear translocation. Proc. Natl. Acad. Sci. USA 2020, 111, 3068–3073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Kim, Y.H.; Kwak, M.S.; Lee, B.; Shin, J.M.; Aum, S.; Park, I.H.; Lee, M.G.; Shin, J.S. Secretory autophagy machinery and vesicular trafficking are involved in HMGB1 secretion. Autophagy 2020, 1–18. [Google Scholar] [CrossRef] [PubMed]
  126. Yang, H.; Hreggvidsdottir, H.S.; Palmblad, K.; Wang, H.; Ochani, M.; Li, J.; Lu, B.; Chavan, S.; Rosas-Ballina, M.; Al-Abed, Y.; et al. A critical cysteine is required for HMGB1 binding to toll-like receptor 4 and activation of macrophage cytokine release. Proc. Natl. Acad. Sci. USA 2010, 107, 11942–11947. [Google Scholar] [CrossRef] [Green Version]
  127. Kierdorf, K.; Fritz, G. RAGE regulation and signaling in inflammation and beyond. J. Leukoc. Biol. 2013, 94, 55–68. [Google Scholar] [CrossRef] [PubMed]
  128. Sundberg, E.; Fasth, A.E.R.; Palmblad, K.; Harris, H.E.; Andersson, U. High mobility group box chromosomal protein 1 acts as a proliferation signal for activated T lymphocytes. Immunobiology 2009, 214, 303–309. [Google Scholar] [CrossRef]
  129. Harris, H.E.; Andersson, U.; Pisetsky, D.S. HMGB1: A multifunctional alarmin driving autoimmune and inflammatory disease. Nat. Rev. Rheumatol. 2012, 8, 195–202. [Google Scholar] [CrossRef]
  130. Xu, J.; Jiang, Y.; Wang, J.; Shi, X.; Liu, Q.; Liu, Z.; Li, Y.; Scott, M.J.; Xiao, G.; Li, S.; et al. Macrophage endocytosis of high-mobility group box 1 triggers pyroptosis. Cell Death Differ. 2014, 21, 1229–1239. [Google Scholar] [CrossRef] [Green Version]
  131. Manti, S.; Cuppari, C.; Parisi, G.F.; Tardino, L.; Salpietro, C.; Leonardi, S. HMGB1 values and response to HBV vaccine in children with celiac disease. Nutrition 2017, 42, 20–22. [Google Scholar] [CrossRef]
  132. Palone, F.; Vitali, R.; Trovato, C.M.; Montuori, M.; Negroni, A.; Mallardo, S.; Stronati, L. Faecal high mobility group box 1 in children with celiac disease: A pilot study. Dig. Liver Dis. 2018, 50, 916–919. [Google Scholar] [CrossRef]
  133. Piatek-Guziewicz, A.; Ptak-Belowska, A.; Przybylska-Felus, M.; Pasko, P.; Zagrodzki, P.; Brzozowski, T.; Mach, T.; Zwolinska-Wcislo, M. Intestinal parameters of oxidative imbalance in celiac adults with extraintestinal manifestations. World J. Gastroenterol. 2017, 23, 7849–7862. [Google Scholar] [CrossRef]
  134. Hadithi, M.; de Boer, H.; Meijer, J.W.R.; Willekens, F.; Kerckhaert, J.A.; Heijmans, R.; Peña, A.S.; Stehouwer, C.D.A.; Mulder, C.J.J. Coeliac disease in Dutch patients with Hashimoto’s thyroiditis and vice versa. World J. Gastroenterol. 2007, 13, 1715–1722. [Google Scholar] [CrossRef] [Green Version]
  135. Viljamaa, M.; Kaukinen, K.; Huhtala, H.; Kyrönpalo, S.; Rasmussen, M.; Collin, P. Coeliac disease, autoimmune diseases and gluten exposure. Scand. J. Gastroenterol. 2005, 40, 437–443. [Google Scholar] [CrossRef] [PubMed]
  136. Elfström, P.; Sundström, J.; Ludvigsson, J.F. Systematic review with meta-analysis: Associations between coeliac disease and type 1 diabetes. Aliment. Pharmacol. Ther. 2014, 40, 1123–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Zhang, J.; Chen, L.; Wang, F.; Zou, Y.; Li, J.J.; Luo, J.; Khan, F.; Sun, F.; Li, Y.; Liu, J.; et al. Extracellular HMGB1 exacerbates autoimmune progression and recurrence of type 1 diabetes by impairing regulatory T cell stability. Diabetologia 2020, 63, 987–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Li, C.; Peng, S.; Liu, X.; Han, C.; Wang, X.; Jin, T.; Liu, S.; Wang, W.; Xie, X.; He, X.; et al. Glycyrrhizin, a Direct HMGB1 Antagonist, Ameliorates Inflammatory Infiltration in a Model of Autoimmune Thyroiditis via Inhibition of TLR2-HMGB1 Signaling. Thyroid 2017, 27, 722–731. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, R.; Li, H.; Wu, J.; Cai, Z.Y.; Li, B.; Ni, H.; Qiu, X.; Chen, H.; Liu, W.; Yang, Z.H.; et al. Gut stem cell necroptosis by genome instability triggers bowel inflammation. Nature 2020, 580, 386–390. [Google Scholar] [CrossRef] [PubMed]
  140. Messmer, D.; Yang, H.; Telusma, G.; Knoll, F.; Li, J.; Messmer, B.; Tracey, K.J.; Chiorazzi, N. High Mobility Group Box Protein 1: An Endogenous Signal for Dendritic Cell Maturation and Th1 Polarization. J. Immunol. 2004, 173, 307–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Li, G.; Liang, X.; Lotze, M.T. HMGB1: The central cytokine for all lymphoid cells. Front. Immunol. 2013, 4. [Google Scholar] [CrossRef] [Green Version]
  142. Schaper, F.; de Leeuw, K.; Horst, G.; Bootsma, H.; Limburg, P.C.; Heeringa, P.; Bijl, M.; Westra, J. High mobility group box 1 skews macrophage polarization and negatively influences phagocytosis of apoptotic cells. Rheumatology 2016, 55, 2260–2270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Pichery, M.; Mirey, E.; Mercier, P.; Lefrancais, E.; Dujardin, A.; Ortega, N.; Girard, J.-P. Endogenous IL-33 Is Highly Expressed in Mouse Epithelial Barrier Tissues, Lymphoid Organs, Brain, Embryos, and Inflamed Tissues: In Situ Analysis Using a Novel Il-33–LacZ Gene Trap Reporter Strain. J. Immunol. 2012, 188, 3488–3495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Martin, N.T.; Martin, M.U. Interleukin 33 is a guardian of barriers and a local alarmin. Nat. Immunol. 2016, 17, 122–131. [Google Scholar] [CrossRef] [PubMed]
  145. Bessa, J.; Meyer, C.A.; de Vera Mudry, M.C.; Schlicht, S.; Smith, S.H.; Iglesias, A.; Cote-Sierra, J. Altered subcellular localization of IL-33 leads to non-resolving lethal inflammation. J. Autoimmun. 2014, 55, 33–41. [Google Scholar] [CrossRef]
  146. Lüthi, A.U.; Cullen, S.P.; McNeela, E.A.; Duriez, P.J.; Afonina, I.S.; Sheridan, C.; Brumatti, G.; Taylor, R.C.; Kersse, K.; Vandenabeele, P.; et al. Suppression of Interleukin-33 Bioactivity through Proteolysis by Apoptotic Caspases. Immunity 2009, 31, 84–98. [Google Scholar] [CrossRef]
  147. Molofsky, A.B.B.; Savage, A.K.K.; Locksley, R.M.M. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation. Immunity 2015, 42, 1005–1019. [Google Scholar] [CrossRef] [Green Version]
  148. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005, 23, 479–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Saluja, R.; Ketelaar, M.E.; Hawro, T.; Church, M.K.; Maurer, M.; Nawijn, M.C. The role of the IL-33/IL-1RL1 axis in mast cell and basophil activation in allergic disorders. Mol. Immunol. 2015, 63, 80–85. [Google Scholar] [CrossRef]
  150. Salimi, M.; Barlow, J.L.; Saunders, S.P.; Xue, L.; Gutowska-Owsiak, D.; Wang, X.; Huang, L.C.; Johnson, D.; Scanlon, S.T.; McKenzie, A.N.J.; et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J. Exp. Med. 2013, 210, 2939–2950. [Google Scholar] [CrossRef] [PubMed]
  151. Baumann, C.; Bonilla, W.V.; Fröhlich, A.; Helmstetter, C.; Peine, M.; Hegazy, A.N.; Pinschewer, D.D.; Löhning, M. T-bet– and STAT4–dependent IL-33 receptor expression directly promotes antiviral Th1 cell responses. Proc. Natl. Acad. Sci. USA 2015, 112, 4056–4061. [Google Scholar] [CrossRef] [Green Version]
  152. Bonilla, W.V.; Fröhlich, A.; Senn, K.; Kallert, S.; Fernandez, M.; Fallon, P.G.; Klemenz, R.; Nakae, S.; Adler, H.; Merkler, D.; et al. The Alarmin Interleukin-33 Drives. Science 2012, 335, 984–989. [Google Scholar] [CrossRef]
  153. Schiering, C.; Krausgruber, T.; Chomka, A.; Fröhlich, A.; Adelmann, K.; Wohlfert, E.A.; Pott, J.; Griseri, T.; Bollrath, J.; Hegazy, A.N.; et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 2014, 513, 564–568. [Google Scholar] [CrossRef] [Green Version]
  154. Monticelli, L.A.; Osborne, L.C.; Noti, M.; Tran, S.V.; Zaiss, D.M.W.W.; Artis, D. IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin–EGFR interactions. Proc. Natl. Acad. Sci. USA 2015, 112, 10762–10767. [Google Scholar] [CrossRef] [Green Version]
  155. Xiao, Y.; Huang, X.; Zhao, Y.; Chen, F.; Sun, M.; Yang, W.; Chen, L.; Yao, S.; Peniche, A.; Dann, S.M.; et al. Interleukin-33 Promotes REG3γ Expression in Intestinal Epithelial Cells and Regulates Gut Microbiota. Cmgh 2019, 8, 21–36. [Google Scholar] [CrossRef] [Green Version]
  156. Lefrançais, E.; Roga, S.; Gautier, V.; Gonzalez-de-Peredo, A.; Monsarrat, B.; Girard, J.-P.; Cayrol, C. IL-33 is processed into mature bioactive forms by neutrophil elastase and cathepsin G. Proc. Natl. Acad. Sci. USA 2012, 109, 1673–1678. [Google Scholar] [CrossRef] [Green Version]
  157. Lefranҫais, E.; Duval, A.; Mirey, E.; Roga, S.; Espinosa, E.; Cayrol, C.; Girard, J.P. Central domain of IL-33 is cleaved by mast cell proteases for potent activation of group-2 innate lymphoid cells. Proc. Natl. Acad. Sci. USA 2014, 111, 15502–15507. [Google Scholar] [CrossRef] [Green Version]
  158. Villarreal, D.O.; Wise, M.C.; Walters, J.N.; Reuschel, E.L.; Choi, M.J.; Obeng-Adjei, N.; Yan, J.; Morrow, M.P.; Weiner, D.B. Alarmin IL-33 Acts as an immunoadjuvant to enhance antigen-specific tumor immunity. Cancer Res. 2014, 74, 1789–1800. [Google Scholar] [CrossRef] [Green Version]
  159. Bakker, O.B.; Sánchez, A.D.R.; Borek, Z.A.; De Klein, N.; Li, Y.; Modderman, R.; Winkelaar, Y.K.; Johannesen, M.K.; Matarese, F.; Martens, J.H.A.; et al. Potential impact of celiac disease genetic risk factors on T cell receptor signaling in gluten—Specific CD4+ T cells. Sci. Rep. 2021, 11, 1–15. [Google Scholar] [CrossRef] [PubMed]
  160. Benhar, M. Oxidants, antioxidants and thiol Redox switches in the control of regulated cell death pathways. Antioxidants 2020, 9, 309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Jin, X.; Stamnaes, J.; Klöck, C.; DiRaimondo, T.R.; Sollid, L.M.; Khosla, C. Activation of extracellular transglutaminase 2 by thioredoxin. J. Biol. Chem. 2011, 286, 37866–37873. [Google Scholar] [CrossRef] [Green Version]
  162. Plugis, N.M.; Palanski, B.A.; Weng, C.H.; Albertelli, M.; Khosla, C. Thioredoxin-1 selectively activates transglutaminase 2 in the extracellular matrix of the small intestine: Implications for celiac disease. J. Biol. Chem. 2017, 292, 2000–2008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Samir, P.; Malireddi, R.K.S.; Kanneganti, T.D. The PANoptosome: A Deadly Protein Complex Driving Pyroptosis, Apoptosis, and Necroptosis (PANoptosis). Front. Cell. Infect. Microbiol. 2020, 10. [Google Scholar] [CrossRef]
  164. Apelbaum, A.; Yarden, G.; Warszawski, S.; Harari, D.; Schreiber, G. Type I Interferons Induce Apoptosis by Balancing cFLIP and Caspase-8 Independent of Death Ligands. Mol. Cell. Biol. 2013, 33, 800–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Sarhan, J.; Liu, B.C.; Muendlein, H.I.; Weindel, C.G.; Smirnova, I.; Tang, A.Y.; Ilyukha, V.; Sorokin, M.; Buzdin, A.; Fitzgerald, K.A.; et al. Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis. Cell Death Differ. 2019, 26, 332–347. [Google Scholar] [CrossRef] [PubMed]
  166. Therrien, A.; Kelly, C.P.; Silvester, J.A. Celiac Disease: Extraintestinal Manifestations and Associated Conditions. J. Clin. Gastroenterol. 2020, 54, 8–21. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Extrinsic and intrinsic mechanisms of apoptosis playing a role in celiac disease mucosae. On the left side of the picture, there is the scheme of the extrinsic apoptosis mechanism triggered by a CTL. As shown in the picture, in the celiac disease mucosae, the CTLs could trigger apoptosis directly by recognizing stressed-related membrane proteins (MICA, HLA-E) with specific NK receptors (NKG2D, CD94:NKG2C). This event promotes the degranulation of CTLs which release Granzyme B (GZMB) and Perforin-1 (PRF1) in the extracellular medium. PRF1 leads to GZMB entry into the cell cytoplasm which leads to the activation of executioner caspase-3, caspase-8, and MOMP formation. CTLs could also engage death receptor ligands (CD95, TNFα, etc.) to trigger the caspase-8 activation which activates caspase-3. Ultimately, cleaved caspase-3 (CC3) activates the DNA and cytoskeleton protein fragmentation (i.e., dCCK18) and promotes the phosphatidylserine translocation into the outer cell membrane. The last event is recognized as an “eat-me” signal by local phagocytes, which eliminated the apoptotic bodies. The right side of the picture describes different intrinsic apoptotic mechanisms playing a part in celiac disease pathology. Inflammatory cytokines related to CD (IFNγ, TNFα, IFNs type I) and specific gliadin peptides (i.e., p31-43) induce a proapoptotic balance between the BCL-2 family of proteins by increasing pro-apoptotic BAK and repressing BCL-2. This renders a cell prone to induce apoptosis when new stressing conditions appear. Potential stressors are the unfolded protein response (UPR), stressed vesicle traffic (i.e., associated with p31-43 toxicity), and in particular the production of ROS. These stressors induce p53 activation and feed a positive loop back to a proapoptotic BCL-2 family protein balance. If these stressful conditions worsen, the MOMP is formed releasing cytochrome-c into the cytoplasm. This event activates APAF-1 and induces the apoptosome formation which subsequently leads to caspase-3 activation initiating the apoptosis execution phase.
Figure 1. Extrinsic and intrinsic mechanisms of apoptosis playing a role in celiac disease mucosae. On the left side of the picture, there is the scheme of the extrinsic apoptosis mechanism triggered by a CTL. As shown in the picture, in the celiac disease mucosae, the CTLs could trigger apoptosis directly by recognizing stressed-related membrane proteins (MICA, HLA-E) with specific NK receptors (NKG2D, CD94:NKG2C). This event promotes the degranulation of CTLs which release Granzyme B (GZMB) and Perforin-1 (PRF1) in the extracellular medium. PRF1 leads to GZMB entry into the cell cytoplasm which leads to the activation of executioner caspase-3, caspase-8, and MOMP formation. CTLs could also engage death receptor ligands (CD95, TNFα, etc.) to trigger the caspase-8 activation which activates caspase-3. Ultimately, cleaved caspase-3 (CC3) activates the DNA and cytoskeleton protein fragmentation (i.e., dCCK18) and promotes the phosphatidylserine translocation into the outer cell membrane. The last event is recognized as an “eat-me” signal by local phagocytes, which eliminated the apoptotic bodies. The right side of the picture describes different intrinsic apoptotic mechanisms playing a part in celiac disease pathology. Inflammatory cytokines related to CD (IFNγ, TNFα, IFNs type I) and specific gliadin peptides (i.e., p31-43) induce a proapoptotic balance between the BCL-2 family of proteins by increasing pro-apoptotic BAK and repressing BCL-2. This renders a cell prone to induce apoptosis when new stressing conditions appear. Potential stressors are the unfolded protein response (UPR), stressed vesicle traffic (i.e., associated with p31-43 toxicity), and in particular the production of ROS. These stressors induce p53 activation and feed a positive loop back to a proapoptotic BCL-2 family protein balance. If these stressful conditions worsen, the MOMP is formed releasing cytochrome-c into the cytoplasm. This event activates APAF-1 and induces the apoptosome formation which subsequently leads to caspase-3 activation initiating the apoptosis execution phase.
Ijms 22 07426 g001
Figure 2. Pyroptosis a new potential PCD mechanism feeding the inflammatory response in celiac disease patients. IFNs which are upregulated in untreated CD induce the expression of inflammasome sensor components such as AIM2, IFI16, NRLP2, NLRC5, transcription factor IRF1, and cytokine IL-18. At the same time, IRF1, a key factor in CD pathology, can enhance the expression of other inflammasome components whose expression is induced by IL17A and TLRs (caspases-4 and -5, NLRP3 and cytokine IL-1β). This process sensitizes the targeted cells to different inflammasome activating agents. Among them, gliadin peptides, such as p31-43, can activate NLRP3 inflammasome (i.e., p31-43). Additionally, the activation of caspases-4 and -5 could be triggered by potential intracellular LPS release, not currently studied in CD. Ultimately, these events activate caspases-1, -4, and -5 which process pro-IL-1β and pro-IL-18 into their mature bioactive forms. Importantly, during this process, Gasdermin-D is cleaved into N-terminal fragments (GSDMD N-Term) which form oligomers in the cell membrane leading to the release of IL-1β and IL-18 and eventually to pyroptotic cell death. The release of these proteins, as well as other alarmins, activates the local immune cells feeding the local inflammatory response by interacting with its cell receptors (IL-18R and IL-1R).
Figure 2. Pyroptosis a new potential PCD mechanism feeding the inflammatory response in celiac disease patients. IFNs which are upregulated in untreated CD induce the expression of inflammasome sensor components such as AIM2, IFI16, NRLP2, NLRC5, transcription factor IRF1, and cytokine IL-18. At the same time, IRF1, a key factor in CD pathology, can enhance the expression of other inflammasome components whose expression is induced by IL17A and TLRs (caspases-4 and -5, NLRP3 and cytokine IL-1β). This process sensitizes the targeted cells to different inflammasome activating agents. Among them, gliadin peptides, such as p31-43, can activate NLRP3 inflammasome (i.e., p31-43). Additionally, the activation of caspases-4 and -5 could be triggered by potential intracellular LPS release, not currently studied in CD. Ultimately, these events activate caspases-1, -4, and -5 which process pro-IL-1β and pro-IL-18 into their mature bioactive forms. Importantly, during this process, Gasdermin-D is cleaved into N-terminal fragments (GSDMD N-Term) which form oligomers in the cell membrane leading to the release of IL-1β and IL-18 and eventually to pyroptotic cell death. The release of these proteins, as well as other alarmins, activates the local immune cells feeding the local inflammatory response by interacting with its cell receptors (IL-18R and IL-1R).
Ijms 22 07426 g002
Figure 3. Other potential PCD mechanisms in CD pathology. (Top Image): Necroptosis. IFNs are a hallmark of CD patients, and they can stimulate the expression of necroptotic factors such as MLKL and ZBP1 and stimulate the cytoplasmic localization of HMGB1. At the same time, ZBP1 is fundamental for the direct activation of necroptosis by IFNs when caspase-8 is inhibited, by an IFNs/STAT1/ZBP1/RIPK3 mechanism. Additionally, activation of TLRs/TRIF, death receptors (TNFR1, CD95)/TRADD/FADD can induce necroptosis by activating RIPK3 in presence of caspase-8 inhibition. RIPK3 activation leads to MLKL phosphorylation (pMLKL) and migration to the cell membrane in vesicle co-transported with a regulatory factor, ZO-1. As ZO-1 is downregulated in epithelial cells of untreated CD patients, it may indicate a pro-necroptotic phenotype of this cell type. When pMLKL monomers are oligomerized in the plasma membrane, this creates a pore, which leads to necroptosis cell death and the alarmins (IL-1α, IL-33, HMGB1) release. (Bottom Image): Ferroptosis. CD patients show a dramatic increase in ROS production and oxidative stress markers which could be related to a new form of inflammatory PCD called ferroptosis. This PCD could be triggered by inhibiting GPX4 through a reduction of GSH. Both facts have been found in untreated CD patients’ cells. Additionally, IFNγ and p53, both factors related to CD pathology, can reduce the transport of cystine (Cys-Cys) a critical precursor of GSH biosynthesis by inhibiting its membrane transporter (SLC3A2 and SLC7A11). Moreover, the GPX4 downregulation could be a response to the ROS increase if other reductive systems failed (i.e., TRX1). The massive increase in ROS could be triggered by stressed organelles such as mitochondria and the free iron pool. Ultimately, the reduced activity of GPX4 leads to ALOXs, LPCAT3, and ACSL4 increase in membrane PUFA peroxidation and cell death, with the release of alarmins and inflammatory oxidized lipids.
Figure 3. Other potential PCD mechanisms in CD pathology. (Top Image): Necroptosis. IFNs are a hallmark of CD patients, and they can stimulate the expression of necroptotic factors such as MLKL and ZBP1 and stimulate the cytoplasmic localization of HMGB1. At the same time, ZBP1 is fundamental for the direct activation of necroptosis by IFNs when caspase-8 is inhibited, by an IFNs/STAT1/ZBP1/RIPK3 mechanism. Additionally, activation of TLRs/TRIF, death receptors (TNFR1, CD95)/TRADD/FADD can induce necroptosis by activating RIPK3 in presence of caspase-8 inhibition. RIPK3 activation leads to MLKL phosphorylation (pMLKL) and migration to the cell membrane in vesicle co-transported with a regulatory factor, ZO-1. As ZO-1 is downregulated in epithelial cells of untreated CD patients, it may indicate a pro-necroptotic phenotype of this cell type. When pMLKL monomers are oligomerized in the plasma membrane, this creates a pore, which leads to necroptosis cell death and the alarmins (IL-1α, IL-33, HMGB1) release. (Bottom Image): Ferroptosis. CD patients show a dramatic increase in ROS production and oxidative stress markers which could be related to a new form of inflammatory PCD called ferroptosis. This PCD could be triggered by inhibiting GPX4 through a reduction of GSH. Both facts have been found in untreated CD patients’ cells. Additionally, IFNγ and p53, both factors related to CD pathology, can reduce the transport of cystine (Cys-Cys) a critical precursor of GSH biosynthesis by inhibiting its membrane transporter (SLC3A2 and SLC7A11). Moreover, the GPX4 downregulation could be a response to the ROS increase if other reductive systems failed (i.e., TRX1). The massive increase in ROS could be triggered by stressed organelles such as mitochondria and the free iron pool. Ultimately, the reduced activity of GPX4 leads to ALOXs, LPCAT3, and ACSL4 increase in membrane PUFA peroxidation and cell death, with the release of alarmins and inflammatory oxidized lipids.
Ijms 22 07426 g003
Figure 4. A new complex hypothetic scenario for CD pathology. (Top Image): Coexistence of different PCD in CD mucosae. Conclusions. Extrinsic apoptosis is primarily triggered by NK-like CTLs in the epithelium and stressed target cells (MICA+, HLA-E+) in the epithelial compartment. At the same time, inflammatory cytokines released by Th1, CTLs (IFNγ, TNFα), activated dendritic cells and other antigen presenting cells (APC) (Type I IFNs) and activated macrophages (ROS) sensitize different cells, especially epithelial cells, to trigger intrinsic apoptosis. After induction of apoptosis, these cells are detached from the epithelium and are reduced to apoptotic bodies in the lumen. In the end, the excessive death is answered with an increased proliferation rate of transit-amplifying cells. However, the presence of these inflammatory cytokines (IFNγ, TNFα) deteriorates the epithelium capacity to efficiently close the gaps left by apoptotic cells. This may create a series of “epithelial gaps” where microbial components (MAMPs) and gliadin peptides could be introduced in the lumen, triggering a new wave of the inflammatory response by gluten-specific T cells. As previously described, this scenario is appropriate to trigger pyroptosis and necroptosis on sensitized cells. In the end, all the new PCDs could trigger the release of inflammatory factors, such as alarmins and IL-1β and IL-18, which may feed the inflammatory process. (Bottom Image): Potential effect of alarmins and IL-1β/IL-18 in CD mucosae. The different immunogenic factors released from necrotic PCD induce different immune and cell responses. IL-33 and IL-18 have a protective effect on epithelium by increasing epithelial proliferation and AMPs released into the lumen. At the same time, IL-33 has a pro-Th2 and pro-Treg action, which is specifically inhibited by inflammatory cytokines such as IFNg and IL-23. Additionally, IL-1α and IL-1β (IL-1) inhibit Treg phenotypes, potentiating the Th17 polarization and the inflammatory response. Additionally, pro-cytotoxic capacity of CTLs can be enhanced by the presence of IL-33, IL-1, HMGB1 but especially IL-18. Furthermore, IL-1 and HMGB1 enhance the inflammatory capacity of macrophages, but HMGB1 may lead to an eventual pyroptotic cell death acting as a lysosomal LPS carrier. Additionally, HMGB1 promotes T cell proliferation on different T cell populations, and similarly to IL-18, potentiates Th1 polarization during antigen presentation. All this information suggests an exciting future in the study of CD pathology, and the very likely involvement of different PCD mechanisms in the overall pathogeny process and new animal models of CD-triggering events.
Figure 4. A new complex hypothetic scenario for CD pathology. (Top Image): Coexistence of different PCD in CD mucosae. Conclusions. Extrinsic apoptosis is primarily triggered by NK-like CTLs in the epithelium and stressed target cells (MICA+, HLA-E+) in the epithelial compartment. At the same time, inflammatory cytokines released by Th1, CTLs (IFNγ, TNFα), activated dendritic cells and other antigen presenting cells (APC) (Type I IFNs) and activated macrophages (ROS) sensitize different cells, especially epithelial cells, to trigger intrinsic apoptosis. After induction of apoptosis, these cells are detached from the epithelium and are reduced to apoptotic bodies in the lumen. In the end, the excessive death is answered with an increased proliferation rate of transit-amplifying cells. However, the presence of these inflammatory cytokines (IFNγ, TNFα) deteriorates the epithelium capacity to efficiently close the gaps left by apoptotic cells. This may create a series of “epithelial gaps” where microbial components (MAMPs) and gliadin peptides could be introduced in the lumen, triggering a new wave of the inflammatory response by gluten-specific T cells. As previously described, this scenario is appropriate to trigger pyroptosis and necroptosis on sensitized cells. In the end, all the new PCDs could trigger the release of inflammatory factors, such as alarmins and IL-1β and IL-18, which may feed the inflammatory process. (Bottom Image): Potential effect of alarmins and IL-1β/IL-18 in CD mucosae. The different immunogenic factors released from necrotic PCD induce different immune and cell responses. IL-33 and IL-18 have a protective effect on epithelium by increasing epithelial proliferation and AMPs released into the lumen. At the same time, IL-33 has a pro-Th2 and pro-Treg action, which is specifically inhibited by inflammatory cytokines such as IFNg and IL-23. Additionally, IL-1α and IL-1β (IL-1) inhibit Treg phenotypes, potentiating the Th17 polarization and the inflammatory response. Additionally, pro-cytotoxic capacity of CTLs can be enhanced by the presence of IL-33, IL-1, HMGB1 but especially IL-18. Furthermore, IL-1 and HMGB1 enhance the inflammatory capacity of macrophages, but HMGB1 may lead to an eventual pyroptotic cell death acting as a lysosomal LPS carrier. Additionally, HMGB1 promotes T cell proliferation on different T cell populations, and similarly to IL-18, potentiates Th1 polarization during antigen presentation. All this information suggests an exciting future in the study of CD pathology, and the very likely involvement of different PCD mechanisms in the overall pathogeny process and new animal models of CD-triggering events.
Ijms 22 07426 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Perez, F.; Ruera, C.N.; Miculan, E.; Carasi, P.; Chirdo, F.G. Programmed Cell Death in the Small Intestine: Implications for the Pathogenesis of Celiac Disease. Int. J. Mol. Sci. 2021, 22, 7426. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147426

AMA Style

Perez F, Ruera CN, Miculan E, Carasi P, Chirdo FG. Programmed Cell Death in the Small Intestine: Implications for the Pathogenesis of Celiac Disease. International Journal of Molecular Sciences. 2021; 22(14):7426. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147426

Chicago/Turabian Style

Perez, Federico, Carolina Nayme Ruera, Emanuel Miculan, Paula Carasi, and Fernando Gabriel Chirdo. 2021. "Programmed Cell Death in the Small Intestine: Implications for the Pathogenesis of Celiac Disease" International Journal of Molecular Sciences 22, no. 14: 7426. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147426

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