Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
1.
Int J Oncol ; 64(6)2024 06.
Article in English | MEDLINE | ID: mdl-38757345

ABSTRACT

Hepatocellular carcinoma (HCC), one of the leading causes of cancer­related mortality worldwide, is challenging to identify in its early stages and prone to metastasis, and the prognosis of patients with this disease is poor. Treatment options for HCC are limited, with even radical treatments being associated with a risk of recurrence or transformation in the short term. Furthermore, the multi­tyrosine kinase inhibitors approved for first­line therapy have marked drawbacks, including drug resistance and side effects. The rise and breakthrough of immune checkpoint inhibitors (ICIs) have provided a novel direction for HCC immunotherapy but these have the drawback of low response rates. Since avoiding apoptosis is a universal feature of cancer, the induction of non­apoptotic regulatory cell death (NARCD) is a novel strategy for HCC immunotherapy. At present, NARCD pathways, including ferroptosis, pyroptosis and necroptosis, are novel potential forms of immunogenic cell death, which have synergistic effects with antitumor immunity, transforming immune 'cold' tumors into immune 'hot' tumors and exerting antitumor effects. Therefore, these pathways may be targeted as a novel treatment strategy for HCC. In the present review, the roles of ferroptosis, pyroptosis and necroptosis in antitumor immunity in HCC are discussed, and the relevant targets and signaling pathways, and the current status of combined therapy with ICIs are summarized. The prospects of targeting ferroptosis, pyroptosis and necroptosis in HCC immunotherapy are also considered.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Immunotherapy , Liver Neoplasms , Necroptosis , Pyroptosis , Humans , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Pyroptosis/drug effects , Pyroptosis/immunology , Ferroptosis/drug effects , Necroptosis/immunology , Necroptosis/drug effects , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Signal Transduction/drug effects , Animals
2.
Crit Rev Oncol Hematol ; 197: 104340, 2024 May.
Article in English | MEDLINE | ID: mdl-38570176

ABSTRACT

Pyroptosis can be triggered through both canonical and non-canonical inflammasome pathways, involving the cleavage of gasdermin (GSDM) protein family members, like GSDMD and GSDME. The impact of pyroptosis on tumors is nuanced, because its role in regulating cancer progression and anti-tumor immunity may vary depending on the tumor type, stage, location, and immune status. However, pyroptosis cannot be simply categorized as promoting or inhibiting tumors based solely on whether it is acute or chronic in nature. The interplay between pyroptosis and cancer is intricate, with some evidence suggesting that chronic pyroptosis may facilitate tumor growth, while the acute induction of pyroptosis could stimulate anti-cancer immune responses. Tumor hypoxia activates hypoxia inducible factor (HIF) signaling to modulate pyroptosis and immune checkpoint expression. Targeting this hypoxia-pyroptosis-immune escape axis could be a promising therapeutic strategy. This review highlights the complex crosstalk between hypoxia, pyroptosis, and immune evasion in the TME.


Subject(s)
Neoplasms , Pyroptosis , Tumor Escape , Humans , Pyroptosis/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/metabolism , Animals , Tumor Microenvironment/immunology , Signal Transduction , Hypoxia/immunology , Hypoxia/metabolism
3.
Adv Sci (Weinh) ; 11(20): e2305934, 2024 May.
Article in English | MEDLINE | ID: mdl-38484186

ABSTRACT

The Gasdermin protein is a membrane disruptor that can mediate immunogenic pyroptosis and elicit anti-tumor immune function. However, cancer cells downregulate Gasdermin and develop membrane repair mechanisms to resist pyroptosis. Therefore, an artificial membrane disruptor (AMD) that can directly mediate membrane rupture in pyroptosis-deficient cells and induce antitumor immune responses in a controllable manner will be valuable in preclinical and clinical research. A micron-scale Ce6-based AMD that can directly induce plasma membrane rupture (PMR) in gasdermin-deficient tumor cells is established. Micron-scale AMDs localize Ce6 specifically to the plasma membrane without labeling other organelles. Compared to free Ce6 molecules, the use of AMDs results in a higher degree of specificity for the plasma membrane. Due to this specificity, AMDs mediate fast and irreversible PMR under 660 nm red light. Furthermore, the AMDs are capable of inducing programmed cell death and lytic cell death in a catalytic manner, demonstrating that the amount of Ce6 used by AMDs is only one-fifth of that used by Ce6 alone when inducing 80% of cancer cell death. In vivo, the AMDs show specificity for tumor targeting and penetration, suggesting that light-driven programmed cell death is specific to tumors. AMDs are applied to antitumor therapy in gasdermin-deficient tumors, resulting in efficient tumor elimination with minimal damage to major organs when combined with anti-PD-1 therapy. Tumor regression is correlated with PMR-mediated inflammation and T-cell-based immune responses. This study provides new insights for designing bioinspired membrane disruptors for PMR and mediating anti-tumor immunotherapy. Additionally, AMD is a dependable tool for examining the immunogenicity of PMR both in vitro and in vivo.


Subject(s)
Cell Membrane , Animals , Mice , Cell Membrane/metabolism , Humans , Disease Models, Animal , Cell Line, Tumor , Neoplasms/immunology , Pyroptosis/immunology , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism
4.
Front Biosci (Landmark Ed) ; 28(9): 210, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37796716

ABSTRACT

Inflammasomes are cytosolic multi-protein complexes that play an important role in the innate immune system, inducing cytokine maturation and pyroptosis. Trained immunity is the induction of memory in innate immune cells by epigenetic reprogramming due to repeated inflammatory stimuli that alter the inflammatory response and increase resistance to infection or disease. Although it is speculated that nucleotide-binding oligomerization domain (NOD), leucine-rich repeat (LRR), and the NLR family pyrin domain containing 3 (NLRP3) inflammasomes respond to various inflammatory stimuli and are associated with trained immunity, the exact relationship is still unclear. This paper aims to introduce data from recent research on the role of inflammasomes in trained immunity through cellular immunometabolic and epigenetic reprogramming. It also suggests a new therapeutic strategy for inflammatory diseases through the complementary regulation of inflammasomes and trained immunity.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Trained Immunity , Cytokines/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pyroptosis/immunology , Trained Immunity/immunology , Humans , Animals
5.
Biomolecules ; 13(6)2023 05 29.
Article in English | MEDLINE | ID: mdl-37371484

ABSTRACT

BACKGROUND: Understanding the molecular mechanisms driving oncogenic processes in glioma is important in order to develop efficient treatments. Recent studies have proposed gasdermin D (GSDMD) as a newly discovered pyroptosis executive protein associated with tumorigenesis. However, the precise effect of GSDMD on glioma progression remains unknown. METHODS: The expression levels of GSDMD in 931 glioma and 1157 normal control tissues were collected. A series of bioinformatic approaches and in vivo and in vitro experiments were used to investigate the roles and mechanisms of GDSMD in glioma. RESULTS: Significant upregulation of GSDMD was detected in glioma tissues compared to normal brain tissues. Patients with glioma and higher GSDMD levels had shorter overall survival, and the Cox regression analysis revealed that GSDMD was an independent risk factor. In addition, upregulation of GSDMD was associated with higher tumor mutation burden and PD-1/PD-L1 expression. Immune infiltration and single-cell analyses indicated that GSDMD was positively associated with an immunosuppressive microenvironment with more infiltrated macrophages and cancer-associated fibroblasts. Furthermore, the in vitro and in vivo experiments revealed that GSDMD knockdown inhibited glioma proliferation, migration, and growth in vivo. CONCLUSION: Our analyses revealed a relatively comprehensive understanding of the oncogenic role of GSDMD in glioma. GSDMD is a promising prognostic biomarker and a potential target for glioma treatment.


Subject(s)
Gasdermins , Glioma , Tumor Microenvironment , Humans , Gasdermins/genetics , Gasdermins/immunology , Glioma/genetics , Glioma/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Macrophages/immunology , Pyroptosis/genetics , Pyroptosis/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
FEBS J ; 290(6): 1549-1562, 2023 03.
Article in English | MEDLINE | ID: mdl-36181338

ABSTRACT

Sepsis often causes cell death via pyroptosis and hence results in septic cardiomyopathy. Triggering receptors expressed in myeloid cells-1 (TREM-1) may initiate cellular cascade pathways and, in turn, induce cell death and vital organ dysfunction in sepsis, but the evidence is limited. We set to investigate the role of TREM-1 on nucleotide-binding oligomerization domain-like receptors with pyrin domain-3 (NLRP3) inflammasome activation and cardiomyocyte pyroptosis in sepsis models using cardiac cell line (HL-1) and mice. In this study, TREM-1 was found to be significantly increased in HL-1 cells challenged with lipopolysaccharide (LPS). Pyroptosis was also significantly increased in the HL-1 cells challenged with lipopolysaccharide and an NLRP3 inflammasome activator, nigericin. The close interaction between TREM-1 and structural maintenance of chromosome 4 (SMC4) was also identified. Furthermore, inhibition of TREM-1 or SMC4 prevented the upregulation of NLRP3 and decreased Gasdermin-D, IL-1ß and caspase-1 cleavage. In mice subjected to caecal ligation and puncture, the TREM-1 inhibitor LR12 decreased the expression of NLRP3 and attenuated cardiomyocyte pyroptosis, leading to improved cardiac function and prolonged survival of septic mice. Our work demonstrates that, under septic conditions, TREM-1 plays a critical role in cardiomyocyte pyroptosis. Targeting TREM-1 and its associated molecules may therefore lead to novel therapeutic treatments for septic cardiomyopathy.


Subject(s)
Inflammasomes , Myocytes, Cardiac , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Sepsis , Triggering Receptor Expressed on Myeloid Cells-1 , Animals , Humans , Mice , Adenosine Triphosphatases/immunology , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/immunology , Caspase 1/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/immunology , Chromosomes, Human, Pair 4/immunology , Inflammasomes/agonists , Inflammasomes/genetics , Inflammasomes/immunology , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Myeloid Cells/immunology , Myocytes, Cardiac/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pyroptosis/genetics , Pyroptosis/immunology , Sepsis/complications , Sepsis/genetics , Sepsis/immunology , Triggering Receptor Expressed on Myeloid Cells-1/antagonists & inhibitors , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Triggering Receptor Expressed on Myeloid Cells-1/immunology
7.
Yi Chuan ; 45(11): 986-997, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38764264

ABSTRACT

Pyroptosis is a type of programmed cell death mediated by the Gasdermin family. It is triggered in response to pathogen infection or other danger signals. The activation of Gasdermins leads to pyroptosis and the release of large amounts of inflammatory cytokines. Pyroptosis plays a crucial role in combating pathogen infections, as it helps to eliminate infected cells and activate the immune system. However, pathogens have already developed sophisticated strategies to evade or inhibit pyroptosis, allowing them to persist and facilitate infection. This review provides an overview of the discovery of pyroptosis and its importance in anti-infectious immunity. We also discuss several new strategies for inhibiting pyroptosis by pathogens. A thorough learning of the occurrence and regulation of pyroptosis may reveal the pathogenesis of related infectious diseases and contribute to developing effective anti-infective therapeutic strategies.


Subject(s)
Immune Evasion , Pyroptosis , Pyroptosis/immunology , Humans , Animals
8.
Int Immunopharmacol ; 113(Pt A): 109398, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36461597

ABSTRACT

Particulate matter (PM) is a major environmental contaminant that causes and worsens respiratory diseases. Fibroblast growth factor 10 (FGF10), a paracrine fibroblast growth factor that specifically stimulates repair and regeneration after injury, has been shown to protect against PM-induced lung injury. However, the underlying mechanisms are still unclear. In this study, the protective effects of FGF10 were investigated using a PM-induced lung injury mouse model in vivo and BEAS-2B cells in vitro. According to the findings, FGF10 treatment alleviated PM-induced oxidative damage and pyroptosis in vivo and in vitro. Mechanistically, FGF10 activated antioxidative Nrf2 signaling. Inhibition of PI3K signaling with LY294002 or Nrf2 signaling with ML385 revealed that FGF10-mediated lung protection was mediated by the PI3K/Akt/Nrf2 pathway. These results collectively indicate that FGF10 inhibits oxidative stress-mediated pyroptosis via the PI3K/Akt/Nrf2 pathway, suggesting a possible therapy for PM-induced lung injury.


Subject(s)
Fibroblast Growth Factor 10 , Lung Injury , Particulate Matter , Pyroptosis , Animals , Mice , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/immunology , Lung Injury/etiology , Lung Injury/genetics , Lung Injury/immunology , NF-E2-Related Factor 2 , Oxidative Stress/genetics , Oxidative Stress/immunology , Particulate Matter/toxicity , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Pyroptosis/genetics , Pyroptosis/immunology , Signal Transduction
9.
J Ovarian Res ; 15(1): 126, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463191

ABSTRACT

The survival of ovary granulosa cells (GC) is critical in the initiation and progression of polycystic ovary syndrome (PCOS) in females. Here, we found that the PCOS process is accompanied by massive GC pyroptosis resulting from Caspase-1 inflammasome activation. Administration of plumbagin, an effective compound isolated from plant medicine, can prevent the pyroptosis of GC and the onset of PCOS. Mechanistic study indicates the over-activation of the inflammasome in GC is due to the upregulation of WTAP, a key regulator of the RNA N6-methylase complex. WTAP mediates the mRNA N6-methylation of NLRP3 inflammasome component ASC and enhances ASC RNA stability, which results in the overactivation of the inflammasome in GCs from the PCOS model. Plumbagin treatment suppresses the WTAP-mediated N6-methylation of ASC mRNA and reduces the pyroptosis of GCs. This study supports the profound potential of plumbagin in PCOS treatment.


Subject(s)
Granulosa Cells , Naphthoquinones , Polycystic Ovary Syndrome , Pyroptosis , Female , Humans , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Granulosa Cells/drug effects , Granulosa Cells/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Methylation/drug effects , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/immunology , Pyroptosis/drug effects , Pyroptosis/genetics , Pyroptosis/immunology , RNA Splicing Factors/genetics , RNA Splicing Factors/immunology , RNA, Messenger , Naphthoquinones/immunology , Naphthoquinones/pharmacology , Naphthoquinones/therapeutic use
10.
Comput Math Methods Med ; 2022: 1258480, 2022.
Article in English | MEDLINE | ID: mdl-35242200

ABSTRACT

BACKGROUND: Liver hepatocellular carcinoma (LIHC) is a malignance with high incidence and recurrence. Pyroptosis is a programed cell death pattern which both activates the effective immune response and causes cell damage. However, their potential applications of pyroptosis-related genes (PRGs) in the prognostic evaluation and immunotherapy of LIHC are still rarely discussed. METHODS: Comprehensive bioinformatics analyses based on TCGA-LIHC dataset were performed in the current study. RESULTS: A total of 33 PRGs were selected to perform the current study. Of these 33 PRGs, 26 PRGs with upregulation or downregulation in LIHC tissues were identified. We also summarized the related genetic mutation variation landscape. GO and KEGG pathway analysis demonstrated that these 26 PRGs were primarily associated with pyroptosis, positive regulation of interleukin-1 beta production, and NOD-like receptor signaling pathway. An unfavorable OS appeared in LIHC patients with high CASP3, CASP4, CASP6, CASP8, GPX4, GSDMA, GSDME, NLRP3, NLRP7, NOD1, NOD2, PLCG1, and SCAF11 expression and low NLRP6 expression. A prognostic signature constructed by the above 14 prognostic PRGs had moderate to high accuracy to predict LIHC patients' prognosis. And risk score was correlated with the expression of CASP6, CASP8, GPX4, GSDMA, GSDME, NLRP6, and NOD2. Of these 7 genes, CASP8 was identified as the core gene in PPI network. Moreover, lncRNA MIR17HG/hsa-miRNA-130b-3p/CASP8 regulatory axis in LIHC was also detected. CONCLUSIONS: The current study indicated the crucial role of PRGs in the prognostic evaluation of LIHC patients and their correlations with tumor microenvironment in LIHC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Pyroptosis/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Computational Biology , Databases, Genetic/statistics & numerical data , Gene Expression Regulation, Neoplastic , Gene Ontology , Gene Regulatory Networks , Genetic Variation , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Prognosis , Protein Interaction Maps/genetics , Pyroptosis/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Dis Markers ; 2022: 4627845, 2022.
Article in English | MEDLINE | ID: mdl-35308144

ABSTRACT

The importance of cell pyroptosis in immunity regulation is becoming increasingly obvious, especially in diseases of the cardiovascular system. Nevertheless, it is unknown whether the pyroptosis signalling pathway is involved in the immune microenvironment regulation of dilated cardiomyopathy (DCM). Therefore, the purpose of the study was to investigate the influence of pyroptosis on the immune environment in dilated cardiomyopathy. We found that expression of 19 pyrolysis-related genes (PRGs) in DCM samples was altered compared to healthy samples. Subsequently, based on these 12 hub pyrolysis-related genes, we developed a classifier that can distinguish between healthy samples and DCM samples. Among the hub pyrolysis-related genes, RT-PCR analyses demonstrated that five of them exhibited significant differential expression in DCM. Interestingly, we observed that immune characteristics are correlated with pyroptosis: higher expression of GSDMD is positively correlated with infiltrating activated pDCs; GSDMD is negatively correlated with Tregs; CASP1 is positively related to parainflammation; and CASP9 is negatively related to the type II IFN response. In addition, distinct pyroptosis-mediated patterns were identified, and immune characteristics under distinct patterns were revealed: pattern B mediates an active immune response, and pattern A leads to a relatively mild immune response to DCM. We also compared the biological functions between these patterns. Compared with pattern A, pattern B had more abundant pathways, such as the NOTCH signalling pathway and pentose phosphate pathway. In summary, this study proves the important influence of pyrolysis on the immune microenvironment of dilated cardiomyopathy and provides new clues for understanding the pathogenesis of dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated , Gene Expression Profiling , Immunologic Factors , Protein Interaction Maps , Pyroptosis/immunology , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , Databases, Genetic , Datasets as Topic , Humans , Male , Mice , Mice, Inbred C57BL , Signal Transduction
12.
Comput Math Methods Med ; 2022: 3179200, 2022.
Article in English | MEDLINE | ID: mdl-35309841

ABSTRACT

Human immunodeficiency virus (HIV) infection is characterized not only by severe immunodeficiency but also by persistent inflammation and immune activation. These characteristics persist in people living with HIV (PLHIV) receiving effective antiretroviral therapy (ART) and are associated with morbidity and mortality in nonacquired immunodeficiency syndrome (AIDS) events. ART can inhibit HIV replication and promote immune reconstitution, which is currently the most effective way to control AIDS. However, despite effective long-term ART and overall suppression of plasma HIV RNA level, PLHIV still shows chronic low-level inflammation. The exact mechanisms that trigger chronic inflammation are unknown. Activation of the inflammasome is essential for the host response to pathogens, and some recent studies have confirmed the role of the inflammasome in the pathogenesis of inflammatory diseases. The NLRP3 inflammasome has been widely studied, which is a pyrin domain-containing protein 3 belonging to the family of nucleotide-binding and oligomerization domain-like receptors (NLRs). Recent studies suggest that inflammasome-mediated pyroptosis is associated with CD4+ T cell loss in the absence of persistent infectious HIV replication. This article reviews the mechanism of the NLRP3 inflammasome and its correlation with immune reconstitution in PLHIV treated with ART.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Computational Biology , HIV Infections/pathology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Immune Reconstitution , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Pyroptosis/drug effects , Pyroptosis/immunology
13.
Oxid Med Cell Longev ; 2022: 2501279, 2022.
Article in English | MEDLINE | ID: mdl-35132346

ABSTRACT

Eukaryotic cells can initiate several distinct self-destruction mechanisms to display essential roles for the homeostasis maintenance, development, and survival of an organism. Pyroptosis, a key response mode in innate immunity, also referred to as caspase-1-dependent proinflammatory programmed necrotic cell death activated by human caspase-1/4/5, or mouse caspase-1/11, plays indispensable roles in response to cytoplasmic insults and immune defense against infectious diseases. These inflammatory caspases are employed by the host to eliminate pathogen infections such as bacteria, viruses, protozoans, and fungi. Gasdermin D requires to be cleaved and activated by these inflammatory caspases to trigger the pyroptosis process. Physiological rupture of cells results in the release of proinflammatory cytokines, the alarmins IL-1ß and IL-18, symbolizing the inflammatory potential of pyroptosis. Moreover, long noncoding RNAs play direct or indirect roles in the upstream of the pyroptosis trigger pathway. Here, we review in detail recently acquired insights into the central roles of inflammatory caspases, inflammasomes, and pyroptosis, as well as the crosstalk between pyroptosis and long noncoding RNAs in mediating infection immunity and pathogen clearance.


Subject(s)
Caspases/metabolism , Communicable Diseases/immunology , Immunity, Innate , Inflammasomes/metabolism , Pyroptosis/immunology , Signal Transduction/immunology , Alarmins/metabolism , Animals , Communicable Diseases/parasitology , Communicable Diseases/virology , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Humans , Mice , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , RNA, Long Noncoding/metabolism
14.
Scand J Immunol ; 95(2): e13124, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34861056

ABSTRACT

Cells encounter continuous challenges due to tissue insult caused by endogenous and/or exogenous stimuli. Among the mechanisms set in place to counterbalance the tissue insult, innate immunity is always at the forefront. Cells of innate immunity efficiently recognize the 'danger signals' via a specialized set of membrane-bound receptors known as Toll-like receptors. Once this interaction is established, toll-like receptor passes on the responsibility to cytosolic NOD-like receptors through a cascade of signalling pathways. Subsequently, NOD-like receptors assemble to a specialized multiprotein intracellular complex, that is inflammasome. Inflammasome activates Caspase-1 and Gasdermin-D which initiate pyroptotic cell death in the affected tissue by two simultaneous mechanisms. Being a protease, caspase-1 cleaves and activates pro-inflammatory cytokines IL-1ß and IL-18. On the other hand, Gasdermin-D causes proteolytic cleavage which forms a pore in the cell membrane. This review highlights the molecular events ranging from recognition of stimuli to pyroptosis. The review is also an attempt to discuss the mechanisms of the most specific experimental NLRP3 inhibitors.


Subject(s)
Cell Membrane/metabolism , Immunity, Innate/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/immunology , Toll-Like Receptors/metabolism , Alarmins/metabolism , Caspase 1/immunology , Enzyme Inhibitors/pharmacology , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phosphate-Binding Proteins/immunology , Signal Transduction/immunology
15.
JCI Insight ; 6(23)2021 12 08.
Article in English | MEDLINE | ID: mdl-34877938

ABSTRACT

Emerging evidence suggests that astrocyte loss is one of the most important pathological features in the hippocampus of patients with major depressive disorder (MDD) and depressive mice. Pyroptosis is a recently discovered form of programmed cell death depending on Caspase-gasdermin D (Casp-GSDMD), which is involved in multiple neuropsychiatric diseases. However, the involvement of pyroptosis in the onset of MDD and glial pathological injury remains obscure. Here, we observed that depressive mice showed astrocytic pyroptosis, which was responsible for astrocyte loss, and selective serotonin reuptake inhibitor (SSRI) treatment could attenuate the pyroptosis induced by the chronic mild stress (CMS) model. Genetic KO of GSDMD, Casp-1, and astrocytic NOD-like receptor protein 3 (NLRP3) inflammasome in mice alleviated depression-like behaviors and inhibited the pyroptosis-associated protein expression. In contrast, overexpression of astrocytic GSDMD-N-terminal domain (GSDMD-N) in the hippocampus could abolish the improvement of behavioral alterations in GSDMD-deficient mice. This work illustrates that targeting the NLRP3/Casp-1/GSDMD-mediated pyroptosis may provide potential therapeutic benefits to stress-related astrocyte loss in the pathogenesis of depression.


Subject(s)
Astrocytes/metabolism , Caspase 1/metabolism , Depression/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/immunology , Animals , Disease Models, Animal , Humans , Mice
16.
Elife ; 102021 12 23.
Article in English | MEDLINE | ID: mdl-34939932

ABSTRACT

The innate immune system detects pathogens and initiates adaptive immune responses. Inflammasomes are central components of the innate immune system, but whether inflammasomes provide sufficient signals to activate adaptive immunity is unclear. In intestinal epithelial cells (IECs), inflammasomes activate a lytic form of cell death called pyroptosis, leading to epithelial cell expulsion and the release of cytokines. Here, we employed a genetic system to show that simultaneous antigen expression and inflammasome activation specifically in IECs is sufficient to activate CD8+ T cells. By genetic elimination of direct T cell priming by IECs, we found that IEC-derived antigens were cross-presented to CD8+ T cells. However, cross-presentation of IEC-derived antigen to CD8+ T cells only partially depended on IEC pyroptosis. In the absence of inflammasome activation, cross-priming of CD8+ T cells required Batf3+ dendritic cells (conventional type one dendritic cells [cDC1]), whereas cross-priming in the presence of inflammasome activation required a Zbtb46+ but Batf3-independent cDC population. These data suggest the existence of parallel inflammasome-dependent and inflammasome-independent pathways for cross-presentation of IEC-derived antigens.


Subject(s)
Adaptive Immunity/immunology , CD8-Positive T-Lymphocytes/immunology , Inflammasomes/immunology , Intestinal Mucosa/immunology , Animals , Cross-Priming/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Female , Intestinal Mucosa/cytology , Male , Mice , Mice, Transgenic , Pyroptosis/immunology
17.
Cells ; 10(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34943932

ABSTRACT

The extracellular protozoan parasite Giardia duodenalis is a well-known and important causative agent of diarrhea on a global scale. Macrophage pyroptosis has been recognized as an important innate immune effector mechanism against intracellular pathogens. Yet, the effects of noninvasive Giardia infection on macrophage pyroptosis and the associated molecular triggers and regulators remain poorly defined. Here we initially observed that NLRP3 inflammasome-mediated pyroptosis was activated in Giardia-treated macrophages, and inhibition of ROS, NLRP3, or caspase-1 could block GSDMD cleavage, IL-1ß, IL-18 and LDH release, and the cell viability reduction. We also confirmed that Giardia-induced NLRP3 inflammasome activation was involved in its K63 deubiquitination. Thus, six candidate deubiquitinases were screened, among which A20 was identified as an effective regulator. We then screened TLRs on macrophage membranes and found that upon stimulation TLR4 was tightly correlated to ROS enhancement, A20-mediated NLRP3 deubiquitination, and pyroptotic signaling. In addition, several Giardia-secreted proteins were predicted as trigger factors via secretome analysis, of which peptidyl-prolyl cis-trans isomerase B (PPIB) independently induced macrophage pyroptosis. This was similar to the findings from the trophozoite treatment, and also led to the TLR4-mediated activation of NLRP3 through K63 deubiquitination by A20. Collectively, the results of this study have significant implications for expanding our understanding of host defense mechanisms after infection with G. duodenalis.


Subject(s)
Diarrhea/genetics , Giardia lamblia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Animals , Cell Survival/drug effects , Deubiquitinating Enzymes/genetics , Diarrhea/immunology , Diarrhea/parasitology , Disease Models, Animal , Giardia lamblia/immunology , Giardia lamblia/pathogenicity , Host-Parasite Interactions/drug effects , Host-Parasite Interactions/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Inflammasomes/drug effects , Inflammasomes/immunology , Interleukin-18/genetics , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/pharmacology , Macrophages/drug effects , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Peptidylprolyl Isomerase/pharmacology , Phosphate-Binding Proteins/genetics , Pyroptosis/drug effects , Pyroptosis/immunology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/immunology , Trophozoites/drug effects , Trophozoites/pathogenicity , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Ubiquitination/genetics
18.
J Immunol Res ; 2021: 4084566, 2021.
Article in English | MEDLINE | ID: mdl-34734090

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is closely related to glycolipid metabolism and liver inflammation. And there is no effective drug approved for its clinical therapy. In this study, we focused on mangiferin (Man) and explored its effects and mechanisms on NAFLD treatment based on the regulation of glycolipid metabolism and anti-inflammatory in vivo and in vitro. The results exhibited that Man can significantly attenuate liver injury, insulin resistance, and glucose tolerance in high-fat diet- (HFD-) induced NAFLD mice and significantly reduce fat accumulation and inflammation in hepatic tissue of NAFLD mice. The transcriptome level RNA-seq analysis showed that the significantly different expression genes between the Man treatment group and the HFD-induced NAFLD model group were mainly related to regulation of energy, metabolism, and inflammation in liver tissue. Furthermore, western blots, real-time PCR, and immunohistochemistry experiments confirmed that Man significantly activated the AMPK signal pathway and inhibited NLRP3 inflammasome activation and pyroptosis in NAFLD mice. In in vitro cell experiments, we further confirmed that Man can promote glucose consumption and reduce intracellular triglyceride (TG) accumulation induced by free fatty acids in HepG2 cells and further that it can be blocked by AMPK-specific inhibitors. Western blot results showed that Man upregulated p-AMPKα levels and exhibited a significant AMPK activation effect, which was blocked by compound C. At the same time, Man downregulated the expression of NLRP3 inflammasome-related proteins and inhibited the activation of NLRP3 inflammasome, alleviating cell pyroptosis and inflammation effects. These results indicate that Man anti-NAFLD activity is mediated through its regulation of glucolipid metabolism by AMPK activation and its anti-inflammatory effects by NLRP3 inflammasome inhibition. Our study indicates that Man is a promising prodrug for the therapy of NAFLD patients.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Xanthones/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Hep G2 Cells , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Pyroptosis/drug effects , Pyroptosis/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Xanthones/therapeutic use
19.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34730110

ABSTRACT

Impaired wound healing associated with recurrent Staphylococcus aureus infection and unresolved inflammation are hallmarks of nonhealing diabetic foot ulcers (DFUs). Perforin-2, an innate immunity molecule against intracellular bacteria, limits cutaneous infection and dissemination of S. aureus in mice. Here, we report the intracellular accumulation of S. aureus in the epidermis of DFUs with no clinical signs of infection due to marked suppression of perforin-2. S. aureus residing within the epidermis of DFUs triggers AIM2 inflammasome activation and pyroptosis. These findings were corroborated in mice lacking perforin-2. The effects of pyroptosis on DFU clinical outcomes were further elucidated in a 4-week longitudinal clinical study in patients with DFUs receiving standard care. Increased AIM2 inflammasome and ASC-pyroptosome coupled with induction of IL-1ß were found in nonhealing DFUs compared with healing DFUs. Our findings revealed that perforin-2 suppression, intracellular S. aureus accumulation, and associated induction of pyroptosis contribute to healing inhibition and prolonged inflammation in patients with DFUs.


Subject(s)
Diabetic Foot/immunology , Epidermis/immunology , Membrane Proteins/immunology , Pore Forming Cytotoxic Proteins/immunology , Pyroptosis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Wound Healing/immunology , Adult , Aged , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Diabetic Foot/genetics , Diabetic Foot/microbiology , Epidermis/microbiology , Female , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Middle Aged , Pore Forming Cytotoxic Proteins/genetics , Pyroptosis/genetics , Staphylococcal Infections/genetics , Wound Healing/genetics
20.
Eur J Pharmacol ; 913: 174644, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34801532

ABSTRACT

The mortality of sepsis-induced cardiac dysfunction (SICD) is very high due to the complex pathophysiological mechanism. Syringaresinol (SYR) is a natural abstract which possesses anti-inflammatory property. The present study aims was to identify the protective impact of SYR on sepsis-induced cardiac dysfunction and investigate the specific mechanisms. We found that SYR improved the cardiac function and alleviated myocardial injury in mice that subjected to cecal ligation and puncture, in addition, SIRT1 expression was significantly elevated after SYR treatment compared to sepsis group both in vivo and in vitro, along with suppression of NLRP3 activation and proinflammatory cytokines release. However, SIRT1 inhibitor EX427 abolished the impact of SYR on LPS-induced pyroptosis in cardiomyocytes. Furthermore, molecular docking analysis predicted that there is high affinity between SYR and estrogen receptor (ER), ER inhibitor ICI182780, the specific ERß inhibitor PHTP and the specific ERαinhibitor AZD9496 were used to examine the role of ER in the protective effect of SYR against SICD, and the results suggested that ER activation was essential for the cardioprotective function of SYR. In conclusion, SYR ameliorates SICD via the ER/SIRT1/NLRP3/GSDMD pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Furans/pharmacology , Heart Diseases/drug therapy , Lignans/pharmacology , Receptors, Estrogen/metabolism , Sepsis/complications , Animals , Cardiotonic Agents/therapeutic use , Cinnamates/pharmacology , Disease Models, Animal , Fulvestrant/pharmacology , Furans/therapeutic use , Heart/drug effects , Heart Diseases/immunology , Heart Diseases/pathology , Humans , Indoles/pharmacology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Lignans/therapeutic use , Male , Mice , Molecular Docking Simulation , Myocardium/pathology , Pyroptosis/drug effects , Pyroptosis/immunology , Receptors, Estrogen/antagonists & inhibitors , Sepsis/drug therapy , Sepsis/immunology , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...