Abstract
Intestinal homeostasis is achieved by the balance among intestinal epithelium, immune cells, and gut microbiota. Gasdermins (GSDMs), a family of membrane pore forming proteins, can trigger rapid inflammatory cell death in the gut, mainly pyroptosis and NETosis. Importantly, there is increasing literature on the non-cell lytic roles of GSDMs in intestinal homeostasis and disease. While GSDMA is low and PJVK is not expressed in the gut, high GSDMB and GSDMC expression is found restrictively in intestinal epithelial cells. Conversely, GSDMD and GSDME show more ubiquitous expression among various cell types in the gut. The N-terminal region of GSDMs can be liberated for pore formation by an array of proteases in response to pathogen- and danger-associated signals, but it is not fully understood what cell type-specific mechanisms activate intestinal GSDMs. The host relies on GSDMs for pathogen defense, tissue tolerance, and cancerous cell death; however, pro-inflammatory milieu caused by pyroptosis and excessive cytokine release may favor the development and progression of inflammatory bowel disease and cancer. Therefore, a thorough understanding of spatiotemporal mechanisms that control gasdermin expression, activation, and function is essential for the development of future therapeutics for intestinal disorders.
Keywords: GSDM, pyroptosis, intestinal epithelial cells, intestinal bowel disease
Introduction
The first member of the gasdermin family was identified in the upper gastrointestinal tract in 2000 [1]. Up until now, the GSDM family has six members in human, including GSDMA, GSDMB, GSDMC, GSDMD, GSDME (DFNA5) and PJVK (DFNB59, GSDMF) [2]. In mice, GSDMA has three homologues: GSDMA1-3. GSDMB has no homologue in mice. There are four homologues of GSDMC in mice: GSDMC1-4. GSDMD, GSDME, and PJVK each possesses a single homologue in mice. All GSDMs are capable executors of pyroptosis, except for PJVK. While they have conserved N-terminus (NT) and C-terminus (CT), the linker regions differ, which leads to distinct regulations and functions of GSDMs.
GSDMs play pivotal roles in maintaining intestinal homeostasis. They have been shown to induce inflammatory lytic cell death (e.g., pyroptosis and NETosis), rapid cytokine release without cell death, and other non-cell-lytic functions. The aim of this review is to: (1) provide a comprehensive summary of the expression pattern of GSDMs in the intestine based on current literature and single cell RNA sequencing (scRNA-seq) databases; (2) summarize the regulation, cleavage, and functions of GSDMs; and (3) discuss the pathophysiological roles of GSDMs in pathogen-host interactions, inflammatory bowel disease (IBD), and gastrointestinal cancer.
1. Gasdermin Expression in the Intestine
The gut epithelium is a highly regenerative tissue and is replenished every 3-5 days. Considering the translation rate and protein lifetimes, one would predict discordances between mRNA and protein expression in the intestine. Indeed, many genes exhibit mRNA zonated toward the crypt, while proteins are zonated toward the villus, due to delayed protein synthesis [3]. Therefore, caution should be taken when drawing conclusions on intestinal epithelial protein expression and function solely based on mRNA results. For instance, while mouse Gsdmc2-4 mRNA expression is enriched in the transit amplifying (TA) region shown by scRNA-seq and RNAscope, its protein is much more abundant in the villus than the crypt [4]. In addition, we would like to point out that many expression results discussed below were obtained from intestine at homeostatic conditions. Infectious, inflammatory, and tumorigenic environments may vary the expression levels and locations of GSDMs [5].
GSDMs, except PJVK, have been reported or detected at some level in the gastrointestinal (GI) tract. Their expression is mainly found in the epithelium and immune cells, but can also be detected in other cell types including endothelial cells and fibroblasts. In order to decipher the role of individual GSDMs in the intestine, we have to first know which cells express them, at what conditions.
Human GSDMA/ Mouse GSDMA1-3
Although the human GSDMA gene was suggested to be expressed in the upper GI tract and suppressed in gastric cancer [1], further study is needed to determine what subpopulations of human intestinal epithelial cells (IECs), if any, express GSDMA.
Mouse Gsdma genes are primarily expressed in epidermal keratinocytes and hair follicle stem cells [6]. In the mouse intestine, based on scRNA-seq results, Gsdma1 is expressed in a cluster of Chgb+ enteroendocrine cells (EECs), Gsdma2 is expressed in a small subset of goblet cells, while Gsdma3 is barely detectable (Table1, 2) [7, 8].
Table 1.
Expression of human gasdermin genes
| GSDMA | GSDMB | GSDMC | GSDMD | GSDME | PJVK | |
|---|---|---|---|---|---|---|
| Epithelial cells | ||||||
| Stem cells | − | ++ | − | ++++ | − | − |
| Progenitor cells | − | +++ | − | +++ | − | − |
| Transit amplifying cells | − | +++ | − | +++ | − | − |
| Goblet cells | − | ++ | ++ | +++ | + | − |
| Tuft cells | − | − | − | + | − | − |
| Paneth cells | − | + | − | +++ | − | − |
| Microfold cells (M cells) | − | − | − | + | − | − |
| Enterocytes | − | +++ | − | ++++ | ++ | − |
| Enteroendocrine cells (EECs) | − | + | − | ++ | − | − |
| Immune cells | ||||||
| B cells | − | − | − | + | + | − |
| T cells | − | + | − | ++ | − | − |
| Mast cells | − | − | − | + | − | − |
| ILCs | − | − | − | + | − | − |
| Macrophages | − | + | − | ++++ | + | − |
| Dendritic cells (DCs) | − | − | − | +++ | − | − |
| Monocytes | + | − | − | ++++ | ++ | − |
| NK cells | − | − | − | ++ | − | − |
| Other cells | ||||||
| Endothelial cells | − | − | − | +++ | − | − |
| Smooth muscle cells | − | − | − | ++ | − | − |
| Glial cells | − | − | − | ++ | − | − |
| Fibroblasts | − | − | − | +++ | − | − |
| Pericytes | − | − | − | + | − | − |
−: not detected; + : low; ++ : medium; +++ : high; ++++ : very high.
− indicates less than 1% cells express GSDM genes; + indicates 1%-10% cells express low levels of GSDM genes; ++ indicates 10%-30% cells express low or medium levels of GSDM genes; +++ indicates 30%-50% cells express medium or high levels of GSDM genes; ++++ indicates more than 50% cells express high levels of GSDM genes.
The scRNA-seq data is from https://www.ebi.ac.uk/gxa/sc/ and https://singlecell.broadinstitute.org [8]
Table 2.
Expression of mouse gasdermin genes
| Gsdm a1 |
Gsdm a2 |
Gsdm a3 |
Gsdm c1 |
Gsdm c2 |
Gsdm c3 |
Gsdm c4 |
Gsd md |
Gsd me |
Pjv k |
|
|---|---|---|---|---|---|---|---|---|---|---|
| Epithelial cells | ||||||||||
| Stem cells | − | − | − | − | − | − | − | + | − | − |
| Progenit ors | − | − | − | − | ++ | + | ++ | + | − | − |
| Goblet cells | − | + | − | − | + | − | + | + | +++ | − |
| Tuft cells | − | − | − | − | + | − | + | + | − | − |
| Paneth cells | − | − | − | − | − | − | − | + | − | − |
| Enterocyt es | − | − | − | − | +++ | ++ | ++++ | +++ | ++++ | − |
| EECs | + | − | − | − | − | − | − | +++ | ++++ | − |
| Immune cells | ||||||||||
| B cells | − | − | − | − | − | − | − | + | + | − |
| T cells | − | − | − | − | − | − | − | ++ | + | − |
| Mast cells | − | − | − | − | − | − | − | ++++ | +++ | − |
| Macroph ages | − | − | − | − | − | − | − | ++++ | ++ | − |
| Neutrop hils | − | − | − | − | − | − | − | ++ | + | − |
| Dendritic cells | − | − | − | − | − | − | − | ++ | + | − |
| Monocyt es | − | − | − | − | − | − | − | ++++ | + | − |
| NK cells | − | − | − | − | − | − | − | ++ | − | − |
| Other cells | ||||||||||
| Endothelial cells | − | − | − | − | − | − | − | ++ | − | − |
| Smooth muscle | − | − | − | − | − | − | − | ++ | − | − |
| Glial cells | − | − | − | − | − | − | − | ++ | + | − |
| Fibroblas | − | − | − | − | − | − | − | +++ | + | − |
| Pericytes | − | − | − | − | − | − | − | + | + | − |
−: not detected; + : low; ++ : medium; +++ : high; ++++ : very high.
− indicates less than 1% cells express Gsdm genes; + indicates 1%-10% cells express low levels of Gsdm genes; ++ indicates 10%-30% cells express low or medium levels of Gsdm genes; +++ indicates 30%-50% cells express medium or high levels of Gsdm genes; ++++ indicates more than 50% cells express high levels of Gsdm genes.
The scRNA-seq data is from https://bis.zju.edu.cn/MCA/ and https://singlecell.broadinstitute.org [7]
Human GSDMB
The predominant expression site of human GSDMB is the epithelium of lung, esophagus, intestine, and stomach [9-12]. scRNA-seq shows that intestinal GSDMB is expressed in stem cells, TA cells and almost all types of differentiated IECs, expect tuft cells and microfold cells (Table 1). In the immune compartment, GSDMB is expressed by macrophage and T cells [13, 14].
Human GSDMC/ Mouse GSDMC1-4
Human GSDMC expression has been reported to be restricted in the epithelium, not any immune cells, of trachea, esophagus, and intestine [4, 12, 15]. According to human scRNA-seq data, GSDMC high-expressing intestinal cells are goblet cells (Table 1), which is consistent with recent reports in mice [4, 16].
In mice, Gsdmc1 is not expressed in the intestine due to epigenetic silencing (Table 2) [4, 16, 17]. Gsdmc4, Gsdmc2, and Gsdm3 (in the order of their expression levels) can be detected in several types of IECs, including goblet cells, tuft cells, progenitor cells, and enterocytes. It is worth noting that although Gsdmc 2-4 mRNA is enriched in the crypt base and TA region based on sequencing and RNA in situ hybridization [4,16,17], immunostaining revealed that GSDMC protein is only abundant expression in villous enterocytes [4]. Interestingly, the duplicated Gsdmc2-4 genes in mice display an expression pattern analogous to the human-only GSDMB, suggesting that they might be adopted by the two hosts for similar gut functions (see below).
Human GSDMD/Mouse GSDMD
Human GSDMD (hGSDMD) exhibits global expressions in epithelial cells, neurons, cardiomyocytes, and immune cells [2,18]. Almost all cell types in the intestine express some level of hGSDMD, including epithelial cells, fibroblasts, smooth muscle cells, as well as innate and adaptive immune cells (Table 1, 2).
The expression pattern of mouse GSDMD (mGSDMD) is similar to hGSDMD, but some notable differences exist between mice and humans. Mouse GSDMD expression in mast cells is significantly higher than hGSDMD, while in goblet cells, tuft cells, epithelial stem cells and progenitor cells is much lower than that of human. When investigating the function of mGSDMD using mouse models, these expression differences should be considered.
Human GSDME/Mouse GSDME
Human GSDME (hGSDME), also known as DFNA5, was first discovered as a deafness associated gene in the epithelium of cochlea [19]. In the epithelium of intestine, stomach, and esophagus, hGSDME is also highly expressed [20, 21]. In the IECs, human GSDME gene expression is only found in enterocytes and goblet cells (Table 1). Outside of the epithelium, hGSDME is expressed in the endothelium of blood vessels and various immune cells, including B cells, macrophages, and monocytes [2, 22, 23].
Mouse Gsdme is expressed in enterocytes and goblet cells, at a level seemingly higher that of human GSDME, as well as in EECs. Most murine immune cells, including B cells, T cells, mast cells, neutrophils, dendritic cells (DCs), macrophages, and monocytes, express Gsdme. Intriguingly, both Gsdmd and Gsdme are abundantly expressed by mast cells in mice but not in humans, which may contribute to species-specific roles of mast cells in various biological responses. Furthermore, mouse Gsdme is also expressed by glial cells, fibroblasts, and pericytes in mice, implicating their broad and impactful roles.
The cell-type specific expression will provide a new perspective to dissect the roles of GSDMs in the intestine. While gene expression data become more abundantly available, we still lack knowledge of the protein expression and localization of most GSDMs, particularly during disease states. Moreover, the gut is highly compartmentalized in terms of structure (duodenum, jejunum, ileum, and colon), microbial and immune compositions, and function. Many studies on GSDMs only focused on one specific part of the intestine. Spatiotemporal analysis of GSDM distribution and expressional regulation will shed light on their functional roles.
2. Gasdermin cleavage and function
GSDMs play a fundamental role as pore formers of the cell membrane and serve as the executors of pyroptosis [24-26]. Additionally, they are implicated in the promotion of NETosis, a program for formation of neutrophil extracellular traps (NETs) [27, 28]. However, the GSDM pores do not always lead to cell death. Under many conditions, the GSDM pores facilitate the rapid cytokine release and ions fluxes without inducing cell death [4, 29, 30]. The concept of rapid cytokine release was first developed for inflammasome activation in immune cells [31-35]. These pores could be repaired by the endosomal sorting complexes required for transport (ESCRT) or caspase-7 activated acid sphingomyelinase [31, 36]. Furthermore, GSDMs exhibit multifaceted functions beyond pore formation, for example cell proliferation, mucin production, and the maintenance of immune tolerance to food [10, 37, 38].
GSDMA/GSDMA1-3
For cell-lytic function, human GSDMA N-terminus (GSDMANT) has been demonstrated to be a pyroptosis executor in vitro as early as hGSDMD [26]. However, the enzymes that cleave GSDMA in vivo remained unknown for an extended period. Recent studies by Deng et al. and LaRock et al. have shed light on this issue, revealing that Streptococcal pyrogenic exotoxin B (SpeB) from Group A Streptococcus (GAS) cleaves GSDMA and triggers pyroptosis [39, 40] (Figure 1A, B). These results were substantiated through experiments using Gsdma1 KO and Gsdma1-3 KO mice. GAS is a pathogen usually found on the skin and throat. Human GSDMA and mouse Gsdma1-2 may be expressed in specific populations of EECs and goblet cells. It presents an intriguing avenue for research to investigate whether SpeB or other bacterial toxins can cleave GSDMA/GSDMA1-3 in intestinal cells. Until now, the physiological role of hGSDMA/mGSDMA1-3 in the intestine is still unknown and needs further study.
Figure 1. Cleavage of GSDMs.
Human (A) and mouse (B) GSDMs are cleaved in the linker region by microbial and host-derived proteases (listed on top of the proteins in the figure) to release the N-termini (NT), allowing their oligomerization and pore formation on membranes. GSDMB and GSDMD can be inactivated by cleavages in the NT domain by Caspases and viral protease 3C (drawn below the proteins). While GSDMA is not expressed in the intestine, Streptococcal pyrogenc exotoxin B (SpeB) from Group A Streptococcus cleaves GSDMA and triggers pyroptosis. Human GSDMC can be activated by Cas-8 in cancer cells; however, such site is not conserved in all mouse GSDMCs and GSDMC cleavage enzymes in the intestine have not been reported. Differential cleavage events have been reported between human and mouse GSDMD, suggesting species-specific activation and function.
GSDMB
While GSDMB is a well-known asthma related gene, its role in the intestine remains largely unexplored. Recent reports have suggested that GSDMB expressed in epithelial cells can be cleaved at K229 and K244 residues by Granzyme A (GZMA) from cytotoxic CD8+ T cells or NK cells [41, 42] (Figure 1A). The cleaved GSDMB N-terminus triggers pyroptosis and cytokine release [41]. This process can be inhibited by IpaH7.8 effector protein secreted by enteroinvasive Shigella flexneri [42]. It is important to note that different GSDMB isoforms exhibit varying cell-lytic activities. These isoforms are equally targeted by IpaH7.8, but the presence of exon 6 determines their pore-forming activity [43-46]. Notably, some epithelial cell lines used in these reports originate from the intestine, such as Caco2, SW837 and SK-CO-1. However, in vivo evidence supporting the pathophysiological cell-lytic role of GSDMB in response to pathogen invasion is still lacking.
Full-length GSDMB can enhance the activity of Caspase-4 to promote non-canonical pyroptosis in THP1 cells [13]. The functional region mediating this signaling pathway has been identified within the 1-91 amino acids of GSDMB [47]. The activation of apoptotic caspase-7 inhibits non-canonical pyroptosis by cleaving GSDMB at D91 [47, 48] (Figure 1A). After cleavage, the region spanning from GSDMB 92-417 amino acids binds to the 1-91 amino acid sequence and blocks its function. This pathway was confirmed using GSDMB BAC (Bacterial Artifact Chromosome)-transgenic mice that exhibit a severe septic phenotype. The inhibition or deficiency of caspase-7 in GSDMB-transgenic mice exacerbate disease severity [47]. As GSDMB is also expressed in T cells and macrophages in the intestine, albeit at a lower level than in epithelial cells, it is plausible that GSDMB may function as a non-canonical pyroptosis promoter in these immune cells.
GSDMB also exhibits non-cell-lytic function. High levels of GSDMB are found in intestinal epithelial stem cells and TA cells. Recently, Rana et al. reported that GSDMB enhances epithelial restitution in human intestine organoids, and this effect is dependent on PDGF-A-mediated FAK phosphorylation [10] (Figure 2A).
Figure 2. Cell type-specific function of GSDMs in pathogenic infections and intestinal inflammation.
(A) In intestinal epithelial cells (IECs), human full-length GSDMB (BFL) regulates proliferation and restitution for tissue repair. GSDMB N-terminus (BNT) and GSDMD N-terminus (DNT) can directly kill bacteria by forming pores on their membrane. Through not fully understood mechanisms, GSDMC pore (CNT) formation upon helminth infections or IL-10 knockout exacerbates colitis. GSDMD executes diverse functions in IECs by inducing pyroptosis and releasing cytokines to restrain bacterial and viral infections while promoting colitis. Interestingly, a 13-kD fragment of GSDMD (D13KD) promotes CIIA and MHC-II transcription to support Tr1 induction and food tolerance. Activated GSDME (ENT) leads to pyroptosis and facilitates enteroviral pathogenesis and colitis. (B) In goblet cells, cell membrane GSDMCNT and GSDMDNT pores mediate unconventional secretion of IL-33 to expulse helminth and calcium influx for mucin exocytosis, respectively. (C) In macrophages, full-length GSDMB (BFL) activates caspase-4 and −11 to promote non-canonical pyroptosis. GSDMDNT pores promotes cytokine release, PIT and pyroptosis to prevent bacterial pathogens and colitis. (D) In neutrophils, GSDMDNT-dependent pyroptosis facilities while NETosis inhibits bacterial infections. Conversely, GSDMENT-mediated pyroptosis and IL-1β secretion defends Yersinia infection. (E) In innate lymphoid cell type 3 cells, GSDMDNT trigger pyroptosis to defend Salmonella infection in mice.
GSDMC/GSDMC1-4
Human GSDMC can be cleaved by caspase-8 at D240 or D365, under the stimulation of α-KG or hypoxia, respectively (Figure 1A). The cleaved GSDMC N-terminus forms pores on cell membrane and trigger pyroptosis in human breast cancer cells and Hela cells [49, 50]. However, in mice, only the GSDMC4 could be cleaved by caspase-8 at D233 (Figure 1B). This residue is not conserved in mouse GSDMC1-3. Moreover, D365 site is not conserved in murine GSDMCs [50]. In IECs, murine GSDMC2-4 expression is induced by helminth infections [4, 17], which in turn enhances host type 2 immunity for helminth expulsion. While GSDMC cleavage can be detected during helminth infections, protease(s) that cleave intestinal GSDMC in vivo remain enigmatic.
Does GSDMC2-4 activation trigger intestinal pyroptosis during helminth infections? While Xi et al. reported that murine GSDMC2-4 expression is induced by IL-4 and IL-13 [17], type 2 cytokines have never been shown to induce cell death and thus their reported lytic cell death requires a greater scrutiny. On the other hand, Zhao et al. demonstrated that murine GSDMCs undergo cleavage and membrane localization during helminth infections using a validated antibody [4]. Activated by type 2 cytokines and STAT6 signaling, GSDMC pores serve as a conduit for unconventional IL-33 secretion in goblet cells (Figure 2B), without significant lytic cell death [4]. It is worth noting that murine GSDMCs is expressed in all villous IECs besides goblet cells, suggesting that GSDMC proteins may have other yet-to-be defined, non-lytic functions in addition to IL-33 release.
Recently, Du et al. found that Lgr5+ stem cell apoptosis and premature death of mice resulted from Mettl14 depletion are associated with loss of Gsdmc2-4 [51]. GSDMC RNAi knockdown resulted in apoptosis in human colonic organoids. However, these results are counterintuitive and could be off-target effects of RNAi experiments. Instead, independently generated whole-body or IEC-specific Gsdmc1-4 KO mice develop normally [4, 52], suggesting that targets other than Gsdmc genes mediate Mettl14’s function on stem cell survival. Finally, human GSDMC is very low expressed in the intestine and its regulation, cleavage, and function are almost completely unknown.
Human GSDMD/Mouse GSDMD
GSDMD is the most broadly expressed and well-studied member of the GSDM family. It can be cleaved by various enzymes, generating different pore-forming N-termini (Figure 1). In canonical and non-canonical pyroptosis, hGSDMD is cleaved at D275 by caspase-1 and caspase-4/5 (mGSDMD is cleaved at D275 by caspase-1 and caspase-11), respectively, and then becomes the most prominent executor of pyroptosis [24-26]. When infected by Yersinia, caspase-8 cleaves mGSDMD and induces pyroptosis in macrophages [23, 53]. mGSDMD in neutrophils and monocytes can be cleaved by cathepsin G at L274 [54]. mGSDMD in neutrophils can also be cleaved the neutrophil elastase at V251 site [27, 55]. These cleaved GSDMD N-terminus form pores mainly on the cell membrane, as well as on the membrane of other organelles, like mitochondria, azurophilic granules, and autophagosomes [56-58]. Conversely, activated caspase-3 and -7 can cleave hGSDMD at D87 site (mGSDMD at D88) to generate a non-pore-forming 13kD N-terminus [59, 60]. This apoptotic cleavage of hGSDMD/mGSDMD blocks pyroptosis in vitro [59, 60], but is not potent enough to inhibit pyroptosis in vivo [61].
Although mGSDMD is dispensable for intestinal epithelium development [12, 62], it plays a multifaceted role in homeostatic responses to external stimulations. mGSDMD in goblet cells promotes mucin granule exocytosis through calcium influx-mediated F-actin disassembly [63], thus protecting the host from microbial and pathogen invasion (Figure 2B). Recently, He et al. found that dietary antigens activate Caspase-3 and -7 to generate the apoptotic 13KD mGSDMD fragment (mGSDMD13kD) in duodenal IECs. mGSDMD13kD then localizes to nucleus and upregulates CIITA and MHCII transcription, leading to Tr1 cell induction and immune tolerance to food antigens [38] (Figure 2A). Furthermore, hGSDMD/mGSDMD N-terminus also form pores on the mitochondrial membrane and promote pyroptosis and necroptosis [64, 65].
Glial cells are closely associated with enteric neurons, and their interaction modulates internal reflexes within the intestine. Enteric neuronal pyroptosis is reported to be a contributing factor to colonic dysmotility induced by a Western diet [66]. Although GSDMD is expressed in glial cells, it is so far unknown if GSDMD is responsible for the pyroptosis of enteric glial cells.
Human GSDME/Mouse GSDME
The pyroptosis induced by GSDME is also called the secondary necrosis. In GSDME highly expressed cells, apoptotic activated caspase-3 cleaves GSDME and leads to pyroptosis prior to apoptosis. Both human and mouse GSDME is cleaved by caspase-3 and −7 at D270 [21, 67] (Figure 1). In addition, granzyme B from cytotoxic cells can cleave hGSDME/mGSDME at the same site [68].
Although GSDME N-terminus forms pores on the cell membrane, it does not always lead to lytic cell death. The hGSDME/mGSDME pores are also important for IL-1β release independent of its pyroptotic ability in macrophages [69]. Furthermore, in TH17 cells stimulated by Candida albicans, hGSDME pores are important for rapid IL-1α release and NLRP3 inflammasome activation [70]. Notably, TH17 cells resist pyroptosis despite hGSDME pores. However, whether GSDME mediates IL-1α secretion in intestinal TH17 requires further investigation. Finally, hGSDME/mGSDME N-terminus not only form pores on the cell membrane, but also permeabilize mitochondria and activate inflammasomes [57].
3. Gasdermins in Microbe-Host Interactions
The intestinal microbiota is a diverse community comprising bacteria, virus and fungi. The interactions among microbiota, IECs and immune cells maintain the intestinal homeostasis. When the physical and chemical barriers formed by IECs are compromised, the commensal microbiota can cause inflammation. The majority of isolated human gut commensal bacterial strains can induce innate immune responses and promote the secretion of cytokines. Particularly, Proteobacteria have been observed to stimulate IL-1β release when co-cultured with innate immune cells [71]. Consistently, mGSDMD targets Proteobacteria in the colon of high-fat diet-feeding mice to protect host from systemic endotoxemia, suggesting GSDMs and pyroptosis may be involved in these immune responses [72].
One of the foremost roles of GSDMs in the intestine is to function as a crucial defense mechanism against pathogen invasions. The studies about Salmonella and Shigella infected mice reveal the roles of GSDMs in the intestines, which is reviewed recently [73]. Although GSDMA is not reported to participate in these processes largely due to its low expression in the intestine, other GSDMs are important for defending various pathogen invasion.
GSDMB
During bacterial infections, GSDMB is cleaved by GZMA from NK cells in IECs. The cleaved N-terminus can target cardiolipin and lipid A, then directly form pores in membranes of Gram-negative bacteria (Figure 2A), such as E.coli, Citrobacter rodentium, Enterobacter cloacae, Shigella and Salmonella. On the other hand, pathogenic microbes are able to target GSDMB as a virulence strategy. For examples, IpaH7.8 effector protein secreted by enteroinvasive Shigella flexneri ubiquitinates GSDMB and inhibits its cleavage [42]. Furthermore, during the infection of E.coli and Salmonella, GSDMB promotes the non-canonical pyroptosis to defend pathogens in macrophages [47] (Figure 2C). Collectively, GSDMB plays an important role in defending bacterial infections.
GSDMC/GSDMC1-4
In mice, Gsdmc genes, not other Gsdm genes, can be drastically induced by parasitic helminths that infect the gut [4, 74]. Type 2 cytokines and STAT6 activation via coordinated phosphorylation and O-GlcNAcylation are absolutely required for the transcriptional upregulation of Gsdmc genes [4]. GSDMC cleavage and cell membrane localization upon helminth infections can be observed in IECs; however, proteases that activate GSDMC remain unidentified. In goblet cells, GSDMCNT pores facilitate the unconventional secretion of IL-33, which is a pleiotropic “alarmin” that amplify type 2 inflammation for effective antihelminth immunity (Figure 2B) [4]. So far, no evidence suggests that GSDMC responds to other pathogens such as bacteria and viruses, indicative the unique role of GSDMC in antihelminth responses.
Human GSDMD/ Mouse GSDMD
Human and mouse GSDMD mediated pyroptosis and rapid cytokine release in innate immune cells and IECs are crucial for the host to defend against various pathogens, including Staphylococcus aureus, E. coli, Salmonella, Francisella, Shigella, Yersinia and rotavirus (Figure 2A)[23, 53, 75-83].
Shiga toxin 2 (Stx2) /LPS complex from pathogenic enterohaemorrhagic E. coli (EHEC) activated non-canonical pyroptosis in human macrophages in vitro (Figure 2C), although this process does not occur in mouse macrophages, as they lack the Stx receptor CD77 [84]. EHEC can also trigger hGSDMD-mediated pyroptosis in IECs. But the pathophysiological function of this process is not yet confirmed [75]. It is important to note that the role of GSDMD in neutrophils differs from that in macrophages (Figure 2D). Kambara et al. reveal that mGSDMD deficiency in neutrophils delays their death and enhanced host defense to E. coli in mice [55].
During Yersinia infection, a bacterial protein named YopJ can block TAK1 and trigger caspase-8 activation. Casapase-8 cleaves mGSDMD and induce pyroptosis in mouse macrophages (Figure 3C), but not in human macrophage [23, 53, 61]. Furthermore, apoptotic caspases cleaved mGSDMD at D88 also promote Yersinia defense [61]. Although mGSDME is also cleaved by caspase-8 during Yersinia infection, loss of mGSDME does not confer further protection [23].
Figure 3. Upregulated GSDMC/Gsdmc gene expression in IBD.
(A) Expression of GSDM genes, determined by RNA-seq, in a prospective cross-sectional cohort consisting of adult IBD patients and controls (GEO accession # GSE193677, n = 498 Crohn's disease, n = 421 UC and 243 controls) [105]. (B) Raw counts (left) and relative expression (right) of mouse Gsdm genes during DSS colitis (GEO accession # GSE131032). (C) Relative expression of mouse Gsdm genes during TNBS colitis (GEO accession # GSE35609).
hGSDMD/mGSDMD in various cell types responds to Salmonella infection. Recently, Fattinger and colleagues reported that only mGSDMD protect mice from acute Salmonella gut infection by using mouse lines lacking individual GSDMs or all GSDMs (Gsdma1-3, Gsdmc1-4, Gsdmd, and Gsdme). The role of GSDMB was not studied since it is not expressed in mice [85]. Zuo et al. reported that mGSDMD-dependent pyroptosis in macrophage protects against infection, which can be counteracted by Salmonella via its plasmid virulence C gene [86]. In IECs, Salmonella infection activates the NAIP-NLRC4 inflammasome, followed by hGSDMD/mGSDMD pore formation for epithelial contractions [77, 87]. However, the expulsion of IECs was also suggested not to depend on hGSDMD/mGSDMD [87-89]. Furthermore, mGSDMD triggers NETosis in neutrophils [27, 28] and pyroptosis in innate lymphoid cell type 3 cells (ILC3s) [90] to defend Salmonella infection in mice (Figure 2D, E). NETosis elicits NETs (Neutrophil Extracellular Traps), those consist of a DNA scaffold adorned with granule-derived proteins, including enzymatically active proteases and antimicrobial peptides. GSDMD-dependent pyroptosis in macrophage participates in the formation of a structure called pore-induced intracellular trap (PIT), which is different from NETs.
mGSDMD-dependent pyroptosis in macrophage participates in the formation of a structure called pore-induced intracellular trap (PIT), which is different from NETs. PIT refers to that the pores on membrane are small enough to retain organelles and bacteria. PIT traps intracellular bacteria and promotes recruitment of and efferocytosis by neutrophils [91]. IL-1β, IL-18, eicosanoids and virulence factors are important for promoting neutrophil recruitment in this process [91-93]. Furthermore, hGSDMD/mGSDMD N-terminus can also directly bind to the membrane of Gram-negative and Gram-positive bacteria, make pores, and kill the bacteria in vitro [76, 94] (Figure 2A, C). However, whether it can be functional during pathogen infection in vivo is not reported.
mGSDMD promotes AIM2 inflammasome activation to protect the host against Francisella novicida, but the downstream of AIM2 is unclear [81]. Nlrp9b is specifically expressed in IECs and rotavirus-infected IECs exhibit Nlrp9b-hGSDMD/mGSDMD dependent pyroptosis, which helps host restrict rotavirus infection [83]. On the other hand, in Stat1-deficient mice, the Nlrp3/Caspase-1/mGSDMD canonical pyroptosis pathway is immunopathogenic [95].
Human GSDME/Mouse GSDME
hGSDME/mGSDME also protects the host from pathogens. mGSDME is cleaved by apoptotic caspase-3 and activate pyroptosis to induce severe disease during enterovirus 71 (EV71) infection [96] (Figure 2A). mGSDME in neutrophils defends Yersinia infection through inducing pyroptosis and IL-1β release (Figure 2D), but not dependent on NETosis [97]. The cleavage of hGSDME/mGSDME can be activated by TNFα or bacteria toxins, such as cytolethal distending toxin (CDT) from Campylobacter jejuni [98, 99].
To summarize, GSDMs are key regulators of infection (Figure 2). They can directly kill bacteria by pore-forming on bacterial cell membrane and trigger rapid inflammation through host cell pyroptosis and non-cell-lytic cytokine release. They also contribute to the formation of PIT and NET to trap pathogens and disrupt the niche of pathogen replication in IECs. However, an excessive pyroptosis triggered by GSDMs can be detrimental, impeding the host's defense and promoting disease severity during certain viral infections. Therefore, the outcome of host-pathogen interactions can be determined by the location, expression, and activity of GSDMs.
4. Gasdermins in Inflammatory Bowel Disease (IBD)
GSDMs serve as pivotal regulators of intestinal inflammation. The genome-wide association study (GWAS) [100] and data mining efforts [101] have revealed that SNPs of GSDMA, GSDMB, GSDMD and GSDME are associated with IBD. Molecular mechanisms of GSDMs in IBD pathogenesis; however, are incompletely understood.
GSDMA/GSDMA1-3
Although SNPs of GSDMA are associated with IBD, action mechanisms of GSDMA in the intestine remain poorly characterized. Intestinal GSDMA seems to be only expressed by EECs and goblet cells, both of which contribute critically to barrier function and mucosal homeostasis. The GSDMA gene is located near GSDMB on human chromosome 17q12-21, a region highly relevant to asthma and IBD [100, 102]. One plausible hypothesis is that GSDMA SNPs contribute to IBD by regulating GSDMB expression.
GSDMB
GSDMB is one of the most well-known genes associated with autoimmune disease, including asthma and IBD [10, 48, 100, 102-104]. However, mixed results have been reported regarding GSDMB expression by disease variants and in IBD patients [10, 13, 14, 100]. Rana et al. discovered that mutant GSDMB in IBD patients loses the ability to enhance epithelial restitution using cultured human intestine organoids [10] (Figure 2A). Functional characterization of genetic loci linked to GSDMB and whether GSDMB induced immune responses are involved in IBD etiology require more studies.
GSDMC/GSDMC1-4
Although GWAS analyses have not indicated GSDMC’s association with IBD, GSDMC is the only gasdermin gene showing significant upregulation in both ulcerative colitis (UC) and Crohn’s disease (CD) patients (Figure 3A), based on a recent transcriptome analysis on hundreds of intestinal biopsies [105]. In mice, Gsdmc genes (except for the silenced Gsdmc1) not only show higher expression in the intestine than other GSDMs, but also is the only being induced by experimental colitis (Figure 3B, C) [106, 107]. Moreover, mouse GSDMC cleavage is observed in chemically (DSS) and genetically (Il10−/−) induced colitis [4]. Deletion of mouse GSDMCs attenuates DSS colitis exacerbated by helminth infections or IL-10 knockout, suggesting that mouse GSDMC-supported cytokine release contributes to inflammation [4] (Figure 2A). Gsdmc1-4 KO mice showed no significant differences of DSS colitis in naïve wildtype mice, indicating that helminth infection or IL-10 KO induced immune changes facilitate the expression and pro-inflammatory actions of mouse GSDMCs. Further investigations are required to determine the functional involvement of GSDMC in human IBD.
Human GSDMD/Mouse GSDMD
hGSDMD is upregulated in some IBD tissues, especially IECs [108, 109]. Canonical pyroptosis drives experimental colitis in mice. As the executor of canonical pyroptosis, the activation of mGSDMD promotes IL-18 release and colitis [29], while Gsdmd−/− mice exhibit attenuated intestinal inflammation [108]. mGSDMD is also required for IECs FADD deficiency induced ileitis [110] (Figure 2A).
While the prominent role of hGSDMD/mGSDMD in macrophages is to induce pyroptosis and rapid release of IL-1 family cytokines [29, 111], mGSDMD-expressing macrophages in the intestine have been reported to inhibit cGAS-mediated inflammation. As a result, Gsdmd−/− or Gsdmdfl/fl;lyz2cre mice display more severe colitis [112]. We suspect that these conflicting results on Gsdmd−/− mice could be attributed to potential cell-specific roles of mGSDMD – epithelial mGSDMD promotes pyroptosis while macrophage mGSDMD pores mediate potassium efflux to inhibit cGAS-dependent type 1 interferon response [80, 112].
Human GSDME/Mouse GSDME
Epithelial hGSDME/mGSDME is predominant expressed in enterocytes and goblet cells. Tan et al. found that hGSDME participates in the pathogenesis of Crohn’s disease (CD). mGSDME mediates IECs pyroptosis and promotes mucosal inflammation in TNBS-induced colitis [98]. mGSDME KO mice are resistant to intestinal barrier loss [113, 114]. Furthermore, Gsdmd and Gsdme double knockout mice showed almost total resistance to DSS induced colitis [114] (Figure 2A).
As discussed above in section 3 and 4, actions of GSDMs in intestinal homeostasis are intricately balanced. Insufficient GSDM activity permits pathogen invasion, while excessive activation triggers inflammation (Figure 2). Therefore, when investigate the roles of GSDMs in IBD, it is imperative to exercise caution given their double-edged effects.
5. Gasdermins in Intestinal Cancers
Through a pan-cancer analysis, expression of all gasdermin genes has been demonstrated to be extensively perturbed and correlated with patient survival [115]. In colorectal cancer (CRC), GSDMA, GSDMB and GSDMD could negatively regulate cell migration, while GSDME is associated with tumor invasion and metastasis [115, 116]. However, studies of individual GSDM family members in gastrointestinal cancers are still scarce.
GSDMA is a target of LMO1 in TGFβ-dependent apoptotic signaling in gastric cancer, where it is typically suppressed [117]. But it remains undetermined if GSDMA actively contributes to intestinal malignancies.
GZMA cleaved GSDMB induces pyroptosis in cancer cells and promotes the clearance of cancer cells in CRC (Figure 4), suggesting GSDMB can be a tumor suppressor [41].
Figure 4. GSDMs in intestinal tumors and anti-tumor immunity.
In IECs, GSDMC and GSDME can act like oncogenes to promote colorectal cancer development. However, in cancer cells, GSDMB-, C-, D- and E-mediated pyroptotic cell death suppresses tumorigenesis, primarily by activating anti-tumor immunity. Conversely, tumor killing ability of CD8 T cells can be dampened by GSDMD and pyroptosis in dendritic cells and macrophages. Note that GSDMC in driving tumor pyroptosis was observed in breast cancers, not intestinal cancers.
GSDMC was firstly discovered in cancer cells [1, 118]. In a mouse CRC model with simultaneous loss of APC and TGFBR2, Gsdmc2 and Gsdmc4 genes were the most highly regulated [119]. By promoting cell proliferation, GSDMC may function as an oncogene for colorectal carcinogenesis. However, molecular mechanisms driving cancer proliferation remain unknown (Figure 4). In breast cancer, hypoxia facilitates the nuclear translocation of PD-L1, which cooperates with p-STAT3 to transcriptionally activate hGSDMC expression [49]. Subsequently, hGSDMCNT pores are generated by TNFα-activated caspase-8 to induce pyroptotic cell death in breast cancer. Nonetheless, it is unknown if GSDMCNT pores form in intestinal cancers and mediate tumor pyroptosis (Figure 4).
In gastric cancer, low hGSDMD expression promotes cell proliferation through regulating cell cycle-related proteins [120]. In CRC, hGSDMD levels are lower than non-cancerous control tissues [121]. Furthermore, lower hGSDMD levels correlate with poorer prognosis of CRC patients [122]. All these data suggest that hGSDMD suppress intestinal tumorigenesis. However, hGSDMD/mGSDMD induces the cell death of tumor associated antigen presenting cells, such as dendritic cells and macrophage, and impairs the function of CD8+ T cells in tumor [123] (Figure 4). When studying GSDMD in intestinal cancers, cell-specific actions shall be carefully considered.
hGSDME/mGSDME is downregulated in most cancer cell lines [20, 124] and demonstrated to suppress tumor growth by activating anti-tumor immunity [68]. However, in the intestine, mGSDME has been suggested to promote CRC in mice via HMGB1 released from pyroptotic IECs [125] (Figure 4). Surprisingly, Gsdme−/− mice showed no differences compared with wildtype mice in chemically induced and in genetic intestinal cancer mouse models [126], possible due to mGSDME’s dual function in anti-tumorigenesis and pro-inflammation. Cell-specific knockout mouse model will in the future help delineate the precise role of hGSDME/mGSDME in intestinal cancers.
These contrasting results underscore the complex roles of GSDMs in intestinal cancers as well other tumors (Figure 4). No unified mechanism can be generalized to all GSDMs, all cells, or all tumorigenic stages. Just like many other cancer-related pathways, pyroptosis and mediated inflammation can be double-edged swords.
Conclusions and Perspectives
To die or not to die, that is the decision made by GSDMs based on their expression locations and the external or internal stimulations. GSDMs can induce inflammatory cell death of IECs and immune cells to regulate mucosal immunity and anti-tumor immune responses. Meanwhile, GSDM pores allow rapid cytokine and pathogen trap release, as well as intracellular PIT formation, which collectively help host defend pathogenic invasions. However, overactivation of GSDMs leads to inflammatory diseases like IBD and can sometimes promote pathogen infections. Finally, GSDMs non-canonically promote the cell proliferation and gene transcription.
Many open questions remain regarding GSDM in intestinal health and disease. What regulatory mechanisms control GSDM expression in time and space, particularly at the protein level? What proteases activate or inactivate GSDMs in different intestinal cell types in vivo, upon what microbial, immunological, damage, or stress signals? When it comes to pyroptosis and cytokine release, does compensation exist between different GSDM pores? Duplicated Gsdmc2-4 genes in mice demonstrate the similar expression pattern as human GSDMB gene, which was lost in mice. But do mouse GSDMC2-4 function similarly as human GSDMB in the gut? Future studies are needed to address species-specific as well as evolutionarily conserved properties of GSDMs. Answering these questions will provide additional mechanistic insight into GSDM biology and inform therapeutic interventions for intestinal disorders.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Reference:
- 1.Saeki N, et al. , Gasdermin (Gsdm) localizing to mouse Chromosome 11 is predominantly expressed in upper gastrointestinal tract but significantly suppressed in human gastric cancer cells. Mamm Genome, 2000. 11(9): p. 718–24. [DOI] [PubMed] [Google Scholar]
- 2.Broz P, Pelegrín P, and Shao F, The gasdermins, a protein family executing cell death and inflammation. Nature reviews. Immunology, 2020. 20(3): p. 143–157. [DOI] [PubMed] [Google Scholar]
- 3.Harnik Y, et al. , Spatial discordances between mRNAs and proteins in the intestinal epithelium. Nature Metabolism, 2021. 3(12): p. 1680–1693. [DOI] [PubMed] [Google Scholar]
- 4.Zhao M, et al. , Epithelial STAT6 O-GlcNAcylation drives a concerted anti-helminth alarmin response dependent on tuft cell hyperplasia and Gasdermin C. Immunity, 2022. 55(4): p. 623–638.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cadena C, et al. , Epigenetic and transcriptional control of gasdermins. Semin Immunol, 2023. 70: p. 101841. [DOI] [PubMed] [Google Scholar]
- 6.Bai X, et al. , Roles of GasderminA3 in Catagen-Telogen Transition During Hair Cycling. J Invest Dermatol, 2015. 135(9): p. 2162–2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Han X, et al. , Mapping the Mouse Cell Atlas by Microwell-Seq. Cell, 2018. 173(5): p. 1307. [DOI] [PubMed] [Google Scholar]
- 8.Wang Y, et al. , Single-cell transcriptome analysis reveals differential nutrient absorption functions in human intestine. J Exp Med, 2020. 217(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Das S, et al. , GSDMB induces an asthma phenotype characterized by increased airway responsiveness and remodeling without lung inflammation. Proc Natl Acad Sci U S A, 2016. 113(46): p. 13132–13137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rana N, et al. , GSDMB is increased in IBD and regulates epithelial restitution/repair independent of pyroptosis. Cell, 2022. 185(2): p. 283–298 e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Komiyama H, et al. , Alu-derived cis-element regulates tumorigenesis-dependent gastric expression of GASDERMIN B (GSDMB). Genes Genet Syst, 2010. 85(1): p. 75–83. [DOI] [PubMed] [Google Scholar]
- 12.Saeki N, et al. , Distinctive expression and function of four GSDM family genes (GSDMA-D) in normal and malignant upper gastrointestinal epithelium. Genes Chromosomes Cancer, 2009. 48(3): p. 261–71. [DOI] [PubMed] [Google Scholar]
- 13.Chen Q, et al. , GSDMB promotes non-canonical pyroptosis by enhancing caspase-4 activity. J Mol Cell Biol, 2019. 11(6): p. 496–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hu Y, et al. , Autoimmune disease variants regulate GSDMB gene expression in human immune cells and whole blood. Proc Natl Acad Sci U S A, 2017. 114(38): p. E7860–e7862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tamura M, et al. , Members of a novel gene family, Gsdm, are expressed exclusively in the epithelium of the skin and gastrointestinal tract in a highly tissue-specific manner. Genomics, 2007. 89(5): p. 618–29. [DOI] [PubMed] [Google Scholar]
- 16.Nystrom EEL, et al. , An intercrypt subpopulation of goblet cells is essential for colonic mucus barrier function. Science, 2021. 372(6539). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Xi R, et al. , Up-regulation of gasdermin C in mouse small intestine is associated with lytic cell death in enterocytes in worm-induced type 2 immunity. Proc Natl Acad Sci U S A, 2021. 118(30). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shi H, et al. , GSDMD-Mediated Cardiomyocyte Pyroptosis Promotes Myocardial I/R Injury. Circ Res, 2021. 129(3): p. 383–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Maeda Y, et al. , Quantification of TECTA and DFNA5 expression in the developing mouse cochlea. Neuroreport, 2001. 12(15): p. 3223–6. [DOI] [PubMed] [Google Scholar]
- 20.Akino K, et al. , Identification of DFNA5 as a target of epigenetic inactivation in gastric cancer. Cancer Sci, 2007. 98(1): p. 88–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wang Y, et al. , Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature, 2017. 547(7661): p. 99–103. [DOI] [PubMed] [Google Scholar]
- 22.Yao F, et al. , HDAC11 promotes both NLRP3/caspase-1/GSDMD and caspase-3/GSDME pathways causing pyroptosis via ERG in vascular endothelial cells. Cell Death Discov, 2022. 8(1): p. 112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sarhan J, et al. , Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc Natl Acad Sci U S A, 2018. 115(46): p. E10888–e10897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.He WT, et al. , Gasdermin D is an executor of pyroptosis and required for interleukin-1beta secretion. Cell Res, 2015. 25(12): p. 1285–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kayagaki N, et al. , Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature, 2015. 526(7575): p. 666–71. [DOI] [PubMed] [Google Scholar]
- 26.Shi J, et al. , Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature, 2015. 526(7575): p. 660–5. [DOI] [PubMed] [Google Scholar]
- 27.Sollberger G, et al. , Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci Immunol, 2018. 3(26). [DOI] [PubMed] [Google Scholar]
- 28.Chen KW, et al. , Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps. Sci Immunol, 2018. 3(26). [DOI] [PubMed] [Google Scholar]
- 29.Gao H, et al. , Dysregulated Microbiota-Driven Gasdermin D Activation Promotes Colitis Development by Mediating IL-18 Release. Front Immunol, 2021. 12: p. 750841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ruhl S and Broz P, Regulation of Lytic and Non-Lytic Functions of Gasdermin Pores. J Mol Biol, 2022. 434(4): p. 167246. [DOI] [PubMed] [Google Scholar]
- 31.Ruhl S, et al. , ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science, 2018. 362(6417): p. 956–960. [DOI] [PubMed] [Google Scholar]
- 32.Evavold CL, et al. , The Pore-Forming Protein Gasdermin D Regulates Interleukin-1 Secretion from Living Macrophages. Immunity, 2018. 48(1): p. 35–44 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zanoni I, et al. , By Capturing Inflammatory Lipids Released from Dying Cells, the Receptor CD14 Induces Inflammasome-Dependent Phagocyte Hyperactivation. Immunity, 2017. 47(4): p. 697–709 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wolf AJ, et al. , Hexokinase Is an Innate Immune Receptor for the Detection of Bacterial Peptidoglycan. Cell, 2016. 166(3): p. 624–636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zanoni I, et al. , An endogenous caspase-11 ligand elicits interleukin-1 release from living dendritic cells. Science, 2016. 352(6290): p. 1232–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nozaki K, et al. , Caspase-7 activates ASM to repair gasdermin and perforin pores. Nature, 2022. 606(7916): p. 960–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lv J, et al. , Gasdermin E mediates resistance of pancreatic adenocarcinoma to enzymatic digestion through a YBX1-mucin pathway. Nat Cell Biol, 2022. 24(3): p. 364–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.He K, et al. , Gasdermin D licenses MHCII induction to maintain food tolerance in small intestine. Cell, 2023. 186(14): p. 3033–3048 e20. [DOI] [PubMed] [Google Scholar]
- 39.Deng W, et al. , Streptococcal pyrogenic exotoxin B cleaves GSDMA and triggers pyroptosis. Nature, 2022. 602(7897): p. 496–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.LaRock DL, et al. , Group A Streptococcus induces GSDMA-dependent pyroptosis in keratinocytes. Nature, 2022. 605(7910): p. 527–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhou Z, et al. , Granzyme A from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science, 2020. 368(6494). [DOI] [PubMed] [Google Scholar]
- 42.Hansen JM, et al. , Pathogenic ubiquitination of GSDMB inhibits NK cell bactericidal functions. Cell, 2021. 184(12): p. 3178–3191 e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kong Q, et al. , Alternative splicing of GSDMB modulates killer lymphocyte-triggered pyroptosis. Sci Immunol, 2023. 8(82): p. eadg3196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Oltra SS, et al. , Distinct GSDMB protein isoforms and protease cleavage processes differentially control pyroptotic cell death and mitochondrial damage in cancer cells. Cell Death Differ, 2023. 30(5): p. 1366–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang C, et al. , Structural basis for GSDMB pore formation and its targeting by IpaH7.8. Nature, 2023. 616(7957): p. 590–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhong X, et al. , Structural mechanisms for regulation of GSDMB pore-forming activity. Nature, 2023. 616(7957): p. 598–605. [DOI] [PubMed] [Google Scholar]
- 47.Li X, et al. , Apoptotic caspase-7 activation inhibits non-canonical pyroptosis by GSDMB cleavage. Cell Death Differ, 2023. 30(9): p. 2120–2134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chao KL, Kulakova L, and Herzberg O, Gene polymorphism linked to increased asthma and IBD risk alters gasdermin-B structure, a sulfatide and phosphoinositide binding protein. Proc Natl Acad Sci U S A, 2017. 114(7): p. E1128–E1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hou J, et al. , PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang JY, et al. , The metabolite α-KG induces GSDMC-dependent pyroptosis through death receptor 6-activated caspase-8. Cell Res, 2021. 31(9): p. 980–997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Du J, et al. , N(6)-adenomethylation of GsdmC is essential for Lgr5(+) stem cell survival to maintain normal colonic epithelial morphogenesis. Dev Cell, 2022. 57(16): p. 1976–1994 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fattinger SA, et al. , Gasdermin D is the only Gasdermin that provides protection against acute Salmonella gut infection in mice. Proceedings of the National Academy of Sciences, 2023. 120(48): p. e2315503120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Orning P, et al. , Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science, 2018. 362(6418): p. 1064–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Burgener SS, et al. , Cathepsin G Inhibition by Serpinb1 and Serpinb6 Prevents Programmed Necrosis in Neutrophils and Monocytes and Reduces GSDMD-Driven Inflammation. Cell Rep, 2019. 27(12): p. 3646–3656.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kambara H, et al. , Gasdermin D Exerts Anti-inflammatory Effects by Promoting Neutrophil Death. Cell Rep, 2018. 22(11): p. 2924–2936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Aglietti RA, et al. , GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes. Proc Natl Acad Sci U S A, 2016. 113(28): p. 7858–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rogers C, et al. , Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat Commun, 2019. 10(1): p. 1689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Karmakar M, et al. , N-GSDMD trafficking to neutrophil organelles facilitates IL-1beta release independently of plasma membrane pores and pyroptosis. Nat Commun, 2020. 11(1): p. 2212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mai FY, et al. , Caspase-3-mediated GSDME activation contributes to cisplatin- and doxorubicin-induced secondary necrosis in mouse macrophages. Cell Prolif, 2019. 52(5): p. e12663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Taabazuing CY, Okondo MC, and Bachovchin DA, Pyroptosis and Apoptosis Pathways Engage in Bidirectional Crosstalk in Monocytes and Macrophages. Cell Chem Biol, 2017. 24(4): p. 507–514 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Demarco B, et al. , Caspase-8-dependent gasdermin D cleavage promotes antimicrobial defense but confers susceptibility to TNF-induced lethality. Sci Adv, 2020. 6(47). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Fujii T, et al. , Gasdermin D (Gsdmd) is dispensable for mouse intestinal epithelium development. Genesis, 2008. 46(8): p. 418–23. [DOI] [PubMed] [Google Scholar]
- 63.Zhang J, et al. , Epithelial Gasdermin D shapes the host-microbial interface by driving mucus layer formation. Sci Immunol, 2022. 7(68): p. eabk2092. [DOI] [PubMed] [Google Scholar]
- 64.Weindel CG, et al. , Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis. Cell, 2022. 185(17): p. 3214–3231 e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Miao R, et al. , Gasdermin D permeabilization of mitochondrial inner and outer membranes accelerates and enhances pyroptosis. Immunity, 2023. 56(11): p. 2523–2541 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ye L, et al. , Caspase-11-mediated enteric neuronal pyroptosis underlies Western diet-induced colonic dysmotility. J Clin Invest, 2020. 130(7): p. 3621–3636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Rogers C, et al. , Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat Commun, 2017. 8: p. 14128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhang Z, et al. , Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature, 2020. 579(7799): p. 415–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhou B and Abbott DW, Gasdermin E permits interleukin-1 beta release in distinct sublytic and pyroptotic phases. Cell Rep, 2021. 35(2): p. 108998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Chao YY, et al. , Human T(H)17 cells engage gasdermin E pores to release IL-1alpha on NLRP3 inflammasome activation. Nat Immunol, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Spindler MP, et al. , Human gut microbiota stimulate defined innate immune responses that vary from phylum to strain. Cell Host Microbe, 2022. 30(10): p. 1481–1498 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Shi Y, et al. , Host Gasdermin D restrains systemic endotoxemia by capturing Proteobacteria in the colon of high-fat diet-feeding mice. Gut Microbes, 2021. 13(1): p. 1946369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gul E, et al. , Epithelial inflammasomes, gasdermins, and mucosal inflammation - Lessons from Salmonella and Shigella infected mice. Semin Immunol, 2023. 70: p. 101812. [DOI] [PubMed] [Google Scholar]
- 74.Entwistle LJ, et al. , Epithelial-Cell-Derived Phospholipase A(2) Group 1B Is an Endogenous Anthelmintic. Cell Host Microbe, 2017. 22(4): p. 484–493.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhong Q, et al. , Clustering of Tir during enteropathogenic E. coli infection triggers calcium influx-dependent pyroptosis in intestinal epithelial cells. PLoS Biol, 2020. 18(12): p. e3000986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Liu X, et al. , Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature, 2016. 535(7610): p. 153–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Rauch I, et al. , NAIP-NLRC4 Inflammasomes Coordinate Intestinal Epithelial Cell Expulsion with Eicosanoid and IL-18 Release via Activation of Caspase-1 and -8. Immunity, 2017. 46(4): p. 649–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Li Z, et al. , Shigella evades pyroptosis by arginine ADP-riboxanation of caspase-11. Nature, 2021. 599(7884): p. 290–295. [DOI] [PubMed] [Google Scholar]
- 79.Luchetti G, et al. , Shigella ubiquitin ligase IpaH7.8 targets gasdermin D for degradation to prevent pyroptosis and enable infection. Cell Host Microbe, 2021. 29(10): p. 1521–1530 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Banerjee I, et al. , Gasdermin D Restrains Type I Interferon Response to Cytosolic DNA by Disrupting Ionic Homeostasis. Immunity, 2018. 49(3): p. 413–426.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Zhu Q, et al. , Gasdermin D Promotes AIM2 Inflammasome Activation and Is Required for Host Protection against Francisella novicida. J Immunol, 2018. 201(12): p. 3662–3668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Schneider KS, et al. , The Inflammasome Drives GSDMD-Independent Secondary Pyroptosis and IL-1 Release in the Absence of Caspase-1 Protease Activity. Cell Rep, 2017. 21(13): p. 3846–3859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhu S, et al. , Nlrp9b inflammasome restricts rotavirus infection in intestinal epithelial cells. Nature, 2017. 546(7660): p. 667–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Platnich JM, et al. , Shiga Toxin/Lipopolysaccharide Activates Caspase-4 and Gasdermin D to Trigger Mitochondrial Reactive Oxygen Species Upstream of the NLRP3 Inflammasome. Cell Rep, 2018. 25(6): p. 1525–1536.e7. [DOI] [PubMed] [Google Scholar]
- 85.Fattinger SA, et al. , Gasdermin D is the only Gasdermin that provides protection against acute Salmonella gut infection in mice. Proc Natl Acad Sci U S A, 2023. 120(48): p. e2315503120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zuo L, et al. , Salmonella spvC Gene Inhibits Pyroptosis and Intestinal Inflammation to Aggravate Systemic Infection in Mice. Front Microbiol, 2020. 11: p. 562491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Samperio Ventayol P, et al. , Bacterial detection by NAIP/NLRC4 elicits prompt contractions of intestinal epithelial cell layers. Proc Natl Acad Sci U S A, 2021. 118(16). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Sellin ME, et al. , Epithelium-intrinsic NAIP/NLRC4 inflammasome drives infected enterocyte expulsion to restrict Salmonella replication in the intestinal mucosa. Cell Host Microbe, 2014. 16(2): p. 237–248. [DOI] [PubMed] [Google Scholar]
- 89.Knodler LA, et al. , Noncanonical inflammasome activation of caspase-4/caspase-11 mediates epithelial defenses against enteric bacterial pathogens. Cell Host Microbe, 2014. 16(2): p. 249–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Xiong L, et al. , Group 3 innate lymphoid cell pyroptosis represents a host defence mechanism against Salmonella infection. Nat Microbiol, 2022. 7(7): p. 1087–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Jorgensen I, et al. , Pyroptosis triggers pore-induced intracellular traps (PITs) that capture bacteria and lead to their clearance by efferocytosis. J Exp Med, 2016. 213(10): p. 2113–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Jorgensen I, et al. , IL-1beta, IL-18, and eicosanoids promote neutrophil recruitment to pore-induced intracellular traps following pyroptosis. Eur J Immunol, 2016. 46(12): p. 2761–2766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hiyoshi H, et al. , Virulence factors perforate the pathogen-containing vacuole to signal efferocytosis. Cell Host Microbe, 2022. 30(2): p. 163–170 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ding J, et al. , Pore-forming activity and structural autoinhibition of the gasdermin family. Nature, 2016. 535(7610): p. 111–6. [DOI] [PubMed] [Google Scholar]
- 95.Dubois H, et al. , Nlrp3 inflammasome activation and Gasdermin D-driven pyroptosis are immunopathogenic upon gastrointestinal norovirus infection. PLoS Pathog, 2019. 15(4): p. e1007709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Dong S, et al. , Gasdermin E is required for induction of pyroptosis and severe disease during enterovirus 71 infection. J Biol Chem, 2022. 298(5): p. 101850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chen KW, et al. , RIPK1 activates distinct gasdermins in macrophages and neutrophils upon pathogen blockade of innate immune signaling. Proc Natl Acad Sci U S A, 2021. 118(28). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tan G, et al. , Gasdermin-E-mediated pyroptosis participates in the pathogenesis of Crohn's disease by promoting intestinal inflammation. Cell Rep, 2021. 35(11): p. 109265. [DOI] [PubMed] [Google Scholar]
- 99.Gu J, et al. , Campylobacter jejuni Cytolethal Distending Toxin Induces GSDME-Dependent Pyroptosis in Colonic Epithelial Cells. Front Cell Infect Microbiol, 2022. 12: p. 853204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Söderman J, Berglind L, and Almer S, Gene Expression-Genotype Analysis Implicates GSDMA, GSDMB, and LRRC3C as Contributors to Inflammatory Bowel Disease Susceptibility. Biomed Res Int, 2015. 2015: p. 834805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Privitera G, et al. , The gasdermin protein family: emerging roles in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol, 2023. 20(6): p. 366–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hitomi Y, et al. , Identification of the functional variant driving ORMDL3 and GSDMB expression in human chromosome 17q12-21 in primary biliary cholangitis. Sci Rep, 2017. 7(1): p. 2904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Li X, et al. , Genome-wide association studies of asthma indicate opposite immunopathogenesis direction from autoimmune diseases. J Allergy Clin Immunol, 2012. 130(4): p. 861–8.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Christodoulou K, et al. , Next generation exome sequencing of paediatric inflammatory bowel disease patients identifies rare and novel variants in candidate genes. Gut, 2013. 62(7): p. 977–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Argmann C, et al. , Biopsy and blood-based molecular biomarker of inflammation in IBD. Gut, 2023. 72(7): p. 1271–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kremer B, et al. , Temporal colonic gene expression profiling in the recurrent colitis model identifies early and chronic inflammatory processes. PLoS One, 2012. 7(11): p. e50388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Czarnewski P, et al. , Conserved transcriptomic profile between mouse and human colitis allows unsupervised patient stratification. Nat Commun, 2019. 10(1): p. 2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bulek K, et al. , Epithelial-derived gasdermin D mediates nonlytic IL-1beta release during experimental colitis. J Clin Invest, 2020. 130(8): p. 4218–4234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chen X, et al. , NEK7 interacts with NLRP3 to modulate the pyroptosis in inflammatory bowel disease via NF-κB signaling. Cell Death Dis, 2019. 10(12): p. 906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Schwarzer R, et al. , FADD and Caspase-8 Regulate Gut Homeostasis and Inflammation by Controlling MLKL- and GSDMD-Mediated Death of Intestinal Epithelial Cells. Immunity, 2020. 52(6): p. 978–993.e6. [DOI] [PubMed] [Google Scholar]
- 111.Evavold CL, et al. , The Pore-Forming Protein Gasdermin D Regulates Interleukin-1 Secretion from Living Macrophages. Immunity, 2018. 48(1): p. 35–44.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ma C, et al. , Gasdermin D in macrophages restrains colitis by controlling cGAS-mediated inflammation. Sci Adv, 2020. 6(21): p. eaaz6717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Tan G, et al. , An IRF1-dependent Pathway of TNFα-induced Shedding in Intestinal Epithelial Cells. J Crohns Colitis, 2022. 16(1): p. 133–142. [DOI] [PubMed] [Google Scholar]
- 114.Xiao J, et al. , Compound loss of GSDMD and GSDME function is necessary to achieve maximal therapeutic effect in colitis. J Transl Autoimmun, 2022. 5: p. 100162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Mu M, et al. , A pan-cancer analysis of molecular characteristics and oncogenic role of gasdermins. Cancer Cell Int, 2022. 22(1): p. 80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Saeki N, et al. , Distinctive expression and function of four GSDM family genes (GSDMA-D) in normal and malignant upper gastrointestinal epithelium. Genes, chromosomes & cancer, 2009. 48(3): p. 261–71. [DOI] [PubMed] [Google Scholar]
- 117.Saeki N, et al. , GASDERMIN, suppressed frequently in gastric cancer, is a target of LMO1 in TGF-beta-dependent apoptotic signalling. Oncogene, 2007. 26(45): p. 6488–98. [DOI] [PubMed] [Google Scholar]
- 118.Watabe K, et al. , Structure, expression and chromosome mapping of MLZE, a novel gene which is preferentially expressed in metastatic melanoma cells. Jpn J Cancer Res, 2001. 92(2): p. 140–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Miguchi M, et al. , Gasdermin C Is Upregulated by Inactivation of Transforming Growth Factor β Receptor Type II in the Presence of Mutated Apc, Promoting Colorectal Cancer Proliferation. PLOS ONE, 2016. 11(11): p. e0166422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wang WJ, et al. , Downregulation of gasdermin D promotes gastric cancer proliferation by regulating cell cycle-related proteins. J Dig Dis, 2018. 19(2): p. 74–83. [DOI] [PubMed] [Google Scholar]
- 121.Ma Y, et al. , Biological functions and clinical significance of the newly identified long non-coding RNA RP1 −85F18.6 in colorectal cancer. Oncol Rep, 2018. 40(5): p. 2648–2658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wu LS, et al. , LPS Enhances the Chemosensitivity of Oxaliplatin in HT29 Cells via GSDMD-Mediated Pyroptosis. Cancer Manag Res, 2020. 12: p. 10397–10409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Jiang Y, et al. , Gasdermin D restricts anti-tumor immunity during PD-L1 checkpoint blockade. Cell Rep, 2022. 41(4): p. 111553. [DOI] [PubMed] [Google Scholar]
- 124.Masuda Y, et al. , The potential role of DFNA5, a hearing impairment gene, in p53-mediated cellular response to DNA damage. J Hum Genet, 2006. 51(8): p. 652–64. [DOI] [PubMed] [Google Scholar]
- 125.Tan G, et al. , HMGB1 released from GSDME-mediated pyroptotic epithelial cells participates in the tumorigenesis of colitis-associated colorectal cancer through the ERK1/2 pathway. J Hematol Oncol, 2020. 13(1): p. 149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Croes L, et al. , Determination of the Potential Tumor-Suppressive Effects of Gsdme in a Chemically Induced and in a Genetically Modified Intestinal Cancer Mouse Model. Cancers (Basel), 2019. 11(8). [DOI] [PMC free article] [PubMed] [Google Scholar]




