Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cell ; 185(2): 283-298.e17, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35021065

ABSTRACT

Gasdermins are a family of structurally related proteins originally described for their role in pyroptosis. Gasdermin B (GSDMB) is currently the least studied, and while its association with genetic susceptibility to chronic mucosal inflammatory disorders is well established, little is known about its functional relevance during active disease states. Herein, we report increased GSDMB in inflammatory bowel disease, with single-cell analysis identifying epithelial specificity to inflamed colonocytes/crypt top colonocytes. Surprisingly, mechanistic experiments and transcriptome profiling reveal lack of inherent GSDMB-dependent pyroptosis in activated epithelial cells and organoids but instead point to increased proliferation and migration during in vitro wound closure, which arrests in GSDMB-deficient cells that display hyper-adhesiveness and enhanced formation of vinculin-based focal adhesions dependent on PDGF-A-mediated FAK phosphorylation. Importantly, carriage of disease-associated GSDMB SNPs confers functional defects, disrupting epithelial restitution/repair, which, altogether, establishes GSDMB as a critical factor for restoration of epithelial barrier function and the resolution of inflammation.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , Base Sequence , Case-Control Studies , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HEK293 Cells , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Methotrexate/pharmacology , Mutation/genetics , Phosphorylation/drug effects , Polymorphism, Single Nucleotide/genetics , Pyroptosis/drug effects , Pyroptosis/genetics , Reproducibility of Results , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects , Wound Healing/drug effects , Wound Healing/genetics
3.
Proc Natl Acad Sci U S A ; 115(40): E9362-E9370, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30224451

ABSTRACT

Defective and/or delayed wound healing has been implicated in the pathogenesis of several chronic inflammatory disorders, including inflammatory bowel disease (IBD). The resolution of inflammation is particularly important in mucosal organs, such as the gut, where restoration of epithelial barrier function is critical to reestablish homeostasis with the interfacing microenvironment. Although IL-33 and its receptor ST2/ILRL1 are known to be increased and associated with IBD, studies using animal models of colitis to address the mechanism have yielded ambiguous results, suggesting both pathogenic and protective functions. Unlike those previously published studies, we focused on the functional role of IL-33/ST2 during an extended (2-wk) recovery period after initial challenge in dextran sodium sulfate (DSS)-induced colitic mice. Our results show that during acute, resolving colitis the normal function of endogenous IL-33 is protection, and the lack of either IL-33 or ST2 impedes the overall recovery process, while exogenous IL-33 administration during recovery dramatically accelerates epithelial restitution and repair, with concomitant improvement of colonic inflammation. Mechanistically, we show that IL-33 stimulates the expression of a network of microRNAs (miRs) in the Caco2 colonic intestinal epithelial cell (IEC) line, especially miR-320, which is increased by >16-fold in IECs isolated from IL-33-treated vs. vehicle-treated DSS colitic mice. Finally, IL-33-dependent in vitro proliferation and wound closure of Caco-2 IECs is significantly abrogated after specific inhibition of miR-320A. Together, our data indicate that during acute, resolving colitis, IL-33/ST2 plays a crucial role in gut mucosal healing by inducing epithelial-derived miR-320 that promotes epithelial repair/restitution and the resolution of inflammation.


Subject(s)
Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Interleukin-33/metabolism , Intestinal Mucosa/physiology , MicroRNAs/metabolism , Regeneration , Acute Disease , Animals , Caco-2 Cells , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/genetics , Mice , Mice, Knockout , MicroRNAs/genetics
4.
Nat Rev Gastroenterol Hepatol ; 20(6): 366-387, 2023 06.
Article in English | MEDLINE | ID: mdl-36781958

ABSTRACT

Since the identification and characterization of gasdermin (GSDM) D as the main effector of inflammatory regulated cell death (or pyroptosis), literature on the GSDM family of pore-forming proteins is rapidly expanding, revealing novel mechanisms regulating their expression and functions that go beyond pyroptosis. Indeed, a growing body of evidence corroborates the importance of GSDMs within the gastrointestinal system, underscoring their critical contributions to the pathophysiology of gastrointestinal cancers, enteric infections and gut mucosal inflammation, such as inflammatory bowel disease. However, with this increase in knowledge, several important and controversial issues have arisen regarding basic GSDM biology and its role(s) during health and disease states. These include critical questions centred around GSDM-dependent lytic versus non-lytic functions, the biological activities of cleaved versus full-length proteins, the differential roles of GSDM-expressing mucosal immune versus epithelial cells, and whether GSDMs promote pathogenic or protective effects during specific disease settings. This Review provides a comprehensive summary and interpretation of the current literature on GSDM biology, specifically focusing on the gastrointestinal tract, highlighting the main controversial issues and their clinical implications, and addressing future areas of research to unravel the specific role(s) of this intriguing, yet enigmatic, family of proteins.


Subject(s)
Gasdermins , Gastrointestinal Neoplasms , Humans , Neoplasm Proteins/metabolism , Pyroptosis/physiology
5.
Trends Cell Biol ; 33(1): 48-59, 2023 01.
Article in English | MEDLINE | ID: mdl-35821185

ABSTRACT

Gasdermin B (GSDMB) belongs to a family of structurally related proteins [(i.e., gasdermins (GSDMs)]. It distinguishes itself from other members by the lack of autoinhibition but clear bioactivity of its full-length form, its preference to bind to phosphatidylinositol phosphates and sulfatides, and the ability to promote both lytic and nonlytic cellular functions. It is the only gasdermin that lacks a mouse ortholog, making in vivo mechanistic studies challenging to perform. GSDMB is abundantly expressed in epithelial cells lining organs that directly interface with the external environment, such as the gastrointestinal tract, with emerging evidence supporting its role in enteric infections, inflammatory bowel disease (IBD), and colorectal cancer. This review discusses the unique features of GSDMB among other gasdermin family members and controversies surrounding GSDMB-dependent mammalian inflammatory cell death (i.e., pyroptosis), including recent discoveries revealing both lytic and nonlytic functions of epithelial-derived GSDMB, particularly during gut health and disease.


Subject(s)
Gasdermins , Neoplasm Proteins , Animals , Mice , Cell Death , Epithelial Cells/metabolism , Neoplasm Proteins/metabolism , Pyroptosis
6.
Front Cell Infect Microbiol ; 11: 806680, 2021.
Article in English | MEDLINE | ID: mdl-35111698

ABSTRACT

Colorectal cancer (CRC) is one of the most prevalent and deadly forms of cancer in Western countries. Inflammation is a well-known driver of colonic carcinogenesis; however, its role in CRC extends beyond colitis-associated cancer. Over the last decades, numerous associations between intestinal dysbiosis and CRC have been identified, with more recent studies providing mechanistic evidence of a causative relationship. Nonetheless, much remains to be discovered regarding the precise implications of microbiome alterations in the pathogenesis of CRC. Research confirms the importance of a bidirectional crosstalk between the gut microbiome and the mucosal immune system in which inflammasomes, multiprotein complexes that can sense "danger signals," serve as conduits by detecting microbial signals and activating innate immune responses, including the induction of microbicidal activities that can alter microbiome composition. Current evidence strongly supports an active role for this "inflammasome-microbiome axis" in the initiation and development of CRC. Furthermore, the gasdermin (GSDM) family of proteins, which are downstream effectors of the inflammasome that are primarily known for their role in pyroptosis, have been recently linked to CRC pathogenesis. These findings, however, do not come without controversy, as pyroptosis is reported to exert both anti- and protumorigenic functions. Furthermore, the multi-faceted interactions between GSDMs and the gut microbiome, as well as their importance in CRC, have only been superficially investigated. In this review, we summarize the existing literature supporting the importance of the inflammasome-microbiota axis, as well as the activation and function of GSDMs, to gain a better mechanistic understanding of CRC pathogenesis.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Colorectal Neoplasms/pathology , Dysbiosis , Gastrointestinal Microbiome/physiology , Humans , Inflammasomes/metabolism , Pyroptosis
7.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33444291

ABSTRACT

Innate lymphoid cells (ILCs) are enriched at barrier surfaces, including the gastrointestinal tract. While most studies have focused on the balance between pathogenic group 1 ILCs (ILC1s) and protective ILC3s in maintaining gut homeostasis and during chronic intestinal inflammation, such as Crohn's disease (CD), less is known regarding ILC2s. Using an established murine model of CD-like ileitis, i.e., the SAMP1/YitFc (SAMP) mouse strain, we showed that ILC2s, compared with ILC1s and ILC3s, were increased within draining mesenteric lymph nodes and ilea of SAMP versus AKR (parental control) mice early, during the onset of disease. Gut-derived ILC2s from CD patients versus healthy controls were also increased and expanded, similarly to ILC1s, in greater proportion compared with ILC3s. Importantly, we report that the intracellular bacteria-sensing protein, nucleotide-binding oligomerization domaining-containing protein 2, encoded by Nod2, the first and strongest susceptibility gene identified for CD, promoted ILC2 expansion, which was dramatically reduced in SAMP mice lacking NOD2 and in SAMP mice raised under germ-free conditions. Furthermore, these effects occurred through a mechanism involving the IL-33/ST2 ligand-receptor pair. Collectively, our results indicate a functional link between NOD2 and ILC2s, regulated by the IL-33/ST2 axis, that mechanistically may contribute to early events leading to CD pathogenesis.


Subject(s)
Crohn Disease/immunology , Ileitis/immunology , Interleukin-33/immunology , Lymphocytes/immunology , Nod2 Signaling Adaptor Protein/immunology , Signal Transduction/immunology , Animals , Crohn Disease/genetics , Crohn Disease/pathology , Disease Models, Animal , Ileitis/genetics , Ileitis/pathology , Interleukin-33/genetics , Lymphocytes/pathology , Mice , Nod2 Signaling Adaptor Protein/genetics , Signal Transduction/genetics
8.
J Clin Invest ; 130(8): 4218-4234, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32597834

ABSTRACT

Gasdermin D (GSDMD) induces pyroptosis via the pore-forming activity of its N-terminal domain, cleaved by activated caspases associated with the release of IL-1ß. Here, we report a nonpyroptotic role of full-length GSDMD in guiding the release of IL-1ß-containing small extracellular vesicles (sEVs) from intestinal epithelial cells (IECs). In response to caspase-8 inflammasome activation, GSDMD, chaperoned by Cdc37/Hsp90, recruits the E3 ligase, NEDD4, to catalyze polyubiquitination of pro-IL-1ß, serving as a signal for cargo loading into secretory vesicles. GSDMD and IL-1ß colocalize with the exosome markers CD63 and ALIX intracellularly, and GSDMD and NEDD4 are required for release of CD63+ sEVs containing IL-1ß, GSDMD, NEDD4, and caspase-8. Importantly, increased expression of epithelial-derived GSDMD is observed both in patients with inflammatory bowel disease (IBD) and those with experimental colitis. While GSDMD-dependent release of IL-1ß-containing sEVs is detected in cultured colonic explants from colitic mice, GSDMD deficiency substantially attenuates disease severity, implicating GSDMD-mediated release of IL-1ß sEVs in the pathogenesis of intestinal inflammation, such as that observed in IBD.


Subject(s)
Colitis/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/metabolism , Interleukin-1beta/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Animals , Cell Line , Colitis/genetics , Colitis/pathology , Epithelial Cells/pathology , Exosomes/genetics , Exosomes/metabolism , Exosomes/pathology , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-1beta/genetics , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Phosphate-Binding Proteins/genetics , Tetraspanin 30/genetics , Tetraspanin 30/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL