RESUMEN
OBJECTIVE: To investigate the inhibitory effect of bardoxolone methyl (CDDO-Me) on activation of NLRP3 inflammasome and its mechanism for alleviating acute liver injury (ALI). METHODS: Mouse bone marrow-derived macrophages (BMDM) and THP-1 cells were pre-treated with CDDO-Me followed by treatment with Nigericin, ATP, MSU, intracellular LPS transfection for activation of NLRP3 inflammasomes, or poly A: T for activation of AIM2 inflammasomes. The levels of caspase-1 and IL-1ß in the cell culture supernatant was determined with Western blotting and ELISA to assess the inhibitory effect of CDDO-Me on NLRP3 inflammasomes and its specificity. In the animal experiment, male C57BL/6J mouse models of acetaminophen-induced ALI were treated with low-dose (20 mg/kg) and high-dose (40 mg/kg) CDDO-Me, and the changes in serum levels of IL-1ß, TNF- α, AST and ALT were measured by ELISA and liver tissue pathology was observed using HE staining. RESULTS: In mouse BMDM and THP-1 cells, CDDO-Me dose-dependently inhibited the activation of NLRP3 inflammasomes without significantly affecting the secretion of non-inflammasome-related inflammatory factors IL-6 and TNF-α or AIM2 inflammasome activation. In the mouse models of ALI, CDDO-Me treatment at both the low and high doses significantly reduced serum levels of IL-1ß, AST and ALT, ameliorated histological changes and reduced inflammatory cell infiltration in the liver tissue, and the effects exhibited a distinct dose dependence. CONCLUSION: CDDO-Me can specifically inhibit the activation of NLRP3 inflammasomes to alleviate acetaminophen-induced ALI in mice.
Asunto(s)
Inflamasomas , Interleucina-1beta , Proteína con Dominio Pirina 3 de la Familia NLR , Ácido Oleanólico , Animales , Humanos , Masculino , Ratones , Acetaminofén/efectos adversos , Caspasa 1/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Células THP-1 , Factor de Necrosis Tumoral alfa/metabolismoRESUMEN
OBJECTIVES: To observe the effect of electroacupuncture (EA) preconditioning at "Quchi" (LI11) and "Xuehai" (SP10) in prevention of urticaria. METHODS: Twenty-four male SD rats were randomly divided into control, model and preconditioning of EA (Pre-EA) groups (8 rats/group). The urticaria model was established by intradermal injection of dilute allogeneic antioalbumin serum at the spots of the bilateral symmetry of the spine on the back, and followed by tail venous injection of mixture solution of egg albumin diluent, plus 0.5% Evans blue and normal saline. Ten days before the end of modeling, rats of the pre-EA group received EA stimulation of LI11 and SP10 for 20 min, once a day for 10 consecutive days. The times of rat's scratching the sensitized skin were recorded. HE staining method was used to observe the pathological changes of skin tissue, and toluidine blue staining method was used to observe the morphology of mast cells (MCs) in the skin, blood, mesentery, and peritoneal fluid, and calculate the degranulation rate. Immunohistochemical stainning was used to detect immunoglobulin E (IgE), histamine (HIS), and 5-hydroxytryptamine (5-HT) expressions in subcutaneous tissue. NOD like receptor thermal domain associated protein 3 (NLRP3) inflammasome, apoptosis related granule protein (ASC), and cysteine aspartate aminotransferase 1 (Caspase-1) protein expression levels in skin tissue were detected by Western blot. The contents of serum interleukin(IL)-1ß and IL-18 were detected using ELISA method. RESULTS: Compared with the control group, the scratching times, amount of Evans blue exudation of the sensitized blue spots, degranulation rate of MCs in skin, blood, mesentery and peritoneal fluid, the expression levels of IgE, HIS, 5-HT in subcutaneous tissue, protein expression levels of NLRP3, ASC, Caspase-1 in skin tissue, and the contents of serum IL-1ß and IL-18 were significantly increased (P<0.01, P<0.05) in the model group. In comparison with the model group, the scratching times, amount of Evans blue exudation of the sensitized blue spots, degranulation rate of MCs, the expression levels of IgE, HIS, 5-HT in subcutaneous tissue, protein expression levels of NLRP3, ASC, Caspase-1 in skin tissue, and the contents of serum IL-1ß and IL-18 in EA group were significantly decreased (P<0.01, P<0.05). CONCLUSIONS: EA preconditioning at LI11 and SP10 can prevent and treat UR by inhibiting inflammatory response, which is related to the regulation of pyroptosis.
Asunto(s)
Puntos de Acupuntura , Electroacupuntura , Interleucina-1beta , Proteína con Dominio Pirina 3 de la Familia NLR , Ratas Sprague-Dawley , Urticaria , Animales , Masculino , Ratas , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Urticaria/terapia , Urticaria/metabolismo , Inmunoglobulina E/sangre , Mastocitos/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Caspasa 1/metabolismo , Caspasa 1/genética , Histamina/metabolismo , Inflamación/terapia , Inflamación/prevención & control , Inflamación/metabolismo , Inflamación/genéticaRESUMEN
BACKGROUND: Shenghui Yizhi Decoction (SHYZD) has exhibited the capacity to enhance cognitive function and learning abilities in individuals diagnosed with Alzheimer's disease (AD) while ameliorating pre-existing neuroinflammation. Nevertheless, the precise mechanism underlying its therapeutic effects on AD remains to be elucidated. METHODS: Twenty-four male SAMP8 mice were randomly divided into three groups, and eight male SAMR1 mice were used as a blank control, to examine their learning and spatial memory abilities. The expression of amyloid ß1-42 (Aß1-42) was detected by immunohistochemical staining of hippocampal tissue. ELISA was used to detect the interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α) expressions. Real time PCR was used to detect NOD-like receptor thermal protein domain associated protein 3 (NLRP3), cysteine protease-1 (Caspase-1), and IL-1ß mRNA expression. Western blot was used to detect nuclear factor kappa-B (NF-κB), inhibitor of NF-κB α (IκBα), IκB kinase α (IKKα), NLRP3, Caspase-1, and IL-1ß protein expression. RESULTS: In this study, SAMP8 mice, employed as an AD model, displayed markedly diminished abilities in terms of spatial localization, navigation, and spatial exploration when compared to the blank control group. Additionally, there was a substantial upregulation of Aß1-42 expression in the hippocampus of these mice, along with a significant increase in the levels of inflammation-associated factors, including IL-1ß, IL-6, TNF-α, NLRP3, Caspase-1, as well as the NF-κB pathway-related proteins, namely, NF-κB, IκBα, and IKKα. Moreover, after treatment with positive drugs (donepezil hydrochloride) and SHYZD, the learning abilities of the mice exhibited significant improvements. Furthermore, the hallmark AD protein Aß1-42, inflammatory factors, and NF-κB/NLRP3 signalling pathway proteins were significantly reduced. These findings collectively suggest that SHYZD exerts a therapeutic effect on AD. CONCLUSION: In summary, the specific molecular mechanisms through which SHYZD alleviates AD and the potential role for SHYZD in the NF-κB/NLRP3 signalling pathway are identified in this study.
Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Medicamentos Herbarios Chinos , Hipocampo , Transducción de Señal , Animales , Masculino , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Caspasa 1/metabolismo , Caspasa 1/efectos de los fármacos , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Interleucina-1beta/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fragmentos de Péptidos/metabolismo , Transducción de Señal/efectos de los fármacosRESUMEN
There is evidence that in infected cells in vitro the meningococcal HrpA/HrpB two-partner secretion system (TPS) mediates the exit of bacteria from the internalization vacuole and the docking of bacteria to the dynein motor resulting in the induction of pyroptosis. In this study we set out to study the role of the HrpA/HrpB TPS in establishing meningitis and activating pyroptotic pathways in an animal model of meningitis using a reference serogroup C meningococcal strain, 93/4286, and an isogenic hrpB knockout mutant, 93/4286ΩhrpB. Survival experiments confirmed the role of HrpA/HrpB TPS in the invasive meningococcal disease. In fact, the ability of the hrpB mutant to replicate in brain and spread systemically was impaired in mice infected with hrpB mutant. Furthermore, western blot analysis of brain samples during the infection demonstrated that: i. N. meningitidis activated canonical and non-canonical inflammasome pyroptosis pathways in the mouse brain; ii. the activation of caspase-11, caspase-1, and gasdermin-D was markedly reduced in the hrpB mutant; iii. the increase in the amount of IL-1ß and IL-18, which are an important end point of pyroptosis, occurs in the brains of mice infected with the wild-type strain 93/4286 and is strongly reduced in those infected with 93/4286ΩhrpB. In particular, the activation of caspase 11, which is triggered by cytosolic lipopolysaccharide, indicates that during meningococcal infection pyroptosis is induced by intracellular infection after the exit of the bacteria from the internalizing vacuole, a process that is hindered in the hrpB mutant. Overall, these results confirm, in an animal model, that the HrpA/HrpB TPS plays a role in the induction of pyroptosis and suggest a pivotal involvement of pyroptosis in invasive meningococcal disease, paving the way for the use of pyroptosis inhibitors in the adjuvant therapy of the disease.
Asunto(s)
Encéfalo , Caspasa 1 , Modelos Animales de Enfermedad , Meningitis Meningocócica , Neisseria meningitidis , Piroptosis , Animales , Neisseria meningitidis/patogenicidad , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Ratones , Meningitis Meningocócica/microbiología , Caspasa 1/metabolismo , Encéfalo/patología , Encéfalo/microbiología , Encéfalo/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Caspasas/metabolismo , Caspasas Iniciadoras/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas de Unión a Fosfato/genética , Interleucina-1beta/metabolismo , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Sistemas de Secreción Bacterianos/genética , Femenino , Interleucina-18/metabolismo , GasderminasRESUMEN
Tumor immune microenvironment (TIME) spatial organization predicts outcome and therapy response in triple-negative breast cancer (TNBC). An immunosuppressive TIME containing elevated tumor-associated macrophages (TAM) and scarce CD8+ T cells is associated with poor outcome, but the regulatory mechanisms are poorly understood. Here we show that ETS1-driven caspase-1 expression, required for IL1ß processing and TAM recruitment, is negatively regulated by estrogen receptors alpha (ERα) and a defining feature of TNBC. Elevated tumoral caspase-1 is associated with a distinct TIME characterized by increased pro-tumoral TAMs and CD8+ T cell exclusion from tumor nests. Mouse models prove the functional importance of ERα, ETS1, caspase-1 and IL1ß in TIME conformation. Caspase-1 inhibition induces an immunoreactive TIME and reverses resistance to immune checkpoint blockade, identifying a therapeutically targetable mechanism that governs TNBC spatial organization.
Asunto(s)
Caspasa 1 , Inmunoterapia , Neoplasias de la Mama Triple Negativas , Macrófagos Asociados a Tumores , Animales , Femenino , Humanos , Ratones , Caspasa 1/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Interleucina-1beta/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/terapia , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismoRESUMEN
PURPOSE: Alzheimer's disease (AD) is characterized by cognitive decline and abnormal protein accumulation. Copper imbalance and pyroptosis play significant roles in the pathogenesis of AD. Recent studies have suggested that dysregulated copper homeostasis contributed to ß-amyloid accumulation, which may activate the NOD-like receptor protein 3 (NLRP3)-related pyroptosis pathway, promoting neuronal damages and AD progression. Therefore, the present study aims to investigates whether copper facilitates AD through exacerbating ß-amyloid (Aß) induced activation of NLRP3/Caspase-1/Gasdermin D (GSDMD)-mediated neuronal cell pyroptosis. METHODS: Mouse hippocampal HT-22 cells were cultured with Aß1-42 oligomer for 24 h as AD Model group. CuCl2 treatment was administered to the AD cell model, and cell survivability levels were detected by Cell Counting Kit-8 (CCK-8), TdT-mediated dUTP nick end labeling (TUNEL), and other relevant kits. Mitochondrial function was evaluated using Mitochondrial membrane potential dye JC-1 and transmission electron microscopy (TEM). After intervention with the NLRP3 inhibitor MCC950, activation of the NLRP3/Caspase-1/GSDMD pathway by copper ions (Cu2+) was confirmed via Western Blot. Thioredoxin T (ThT) fluorescence assay was performed to observe the aggregation effect of Aß induced by Cu2+ overload. RESULTS: CuCl2 treatment of the AD cell model resulted in up-regulation of the levels of Lactate Dehydrogenase (LDH), Interleukin-1ß (IL-1ß), and IL-18 expression, which indicated activation of pyroptosis. We observed a significant decrease in mitochondrial membrane potential, mitochondrial swelling, and loss of mitochondrial cristae by fluorescence microscopy and TEM. ThT fluorescence imaging showed that Cu2+ promoted Aß aggregation and up-regulated NLRP3, apoptosis-associated speck-like protein containing a CARD (ACS), Caspase-1, Cleaved Caspase-1, GSDMD, and Gasdermin D N-terminal (GSDMD-NT). The NLRP3 inhibitor MCC950 partially reversed Cu2+-mediated pyroptosis in HT-22 cells. CONCLUSIONS: Exposure to copper ions disrupt mitochondrial copper homeostasis, promotes Aß aggregation, and activates NLRP3 inflammasomes, further promoting the Aß aggregation activated pyroptosis in AD cell models.
Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Caspasa 1 , Cobre , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas de Unión a Fosfato , Piroptosis , Piroptosis/fisiología , Piroptosis/efectos de los fármacos , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/farmacología , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Cobre/farmacología , Cobre/metabolismo , Caspasa 1/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Línea Celular , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Sulfonas/farmacología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Indenos/farmacología , Gasderminas , Furanos , SulfonamidasRESUMEN
Localized delivery of inflammasome inhibitors in phagocytic macrophages could be promising for psoriasis treatment. The present work demonstrates the development of non-spherical lipid nanoparticles, mimicking pathogen-like shapes, consisting of an anti-inflammatory inflammasome inhibiting lipid (pyridoxine dipalmitate) as a trojan horse. The nanorods inhibit inflammasome by 3.8- and 4.5-fold compared with nanoellipses and nanospheres, respectively. Nanorods reduce apoptosis-associated speck-like protein and lysosomal rupture, restrain calcium influx, and mitochondrial reactive oxygen species. Dual inflammasome inhibitor (NLRP3/AIM-2-IN-3) loaded nanorods cause synergistic inhibition by 21.5- and 59-folds compared with nanorods and free drug, respectively alongside caspase-1 inhibition. The NLRP3/AIM-2-IN-3 nanorod when transformed into a polymeric scaffold, simultaneously and effectively inhibits RNA levels of NLRP3, AIM2, caspase-1, chemokine ligand-2, gasdermin-D, interleukin-1ß, toll-like receptor 7/ 8, and IL-17A by 6.4-, 1.6-, 2.0-, 13.0-, 4.2-, 24.4-, 4.3-, and 1.82-fold, respectively in psoriatic skin in comparison to Imiquimod positive control group in an in-vivo psoriasis-like mice model.
Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Nanotubos , Psoriasis , Psoriasis/tratamiento farmacológico , Animales , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratones , Humanos , Nanotubos/química , Polímeros/química , Nanopartículas/química , Especies Reactivas de Oxígeno/metabolismo , Modelos Animales de Enfermedad , Lípidos/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Caspasa 1/metabolismo , Femenino , Masculino , LiposomasRESUMEN
Objective To explore the role and potential mechanism of caspase activation and recruitment domain-containing nucleotide-binding oligomerization domain-like receptor 3 (NLRC3) in the immune inflammatory response induced by macrophage pyroptosis in patients with rheumatoid arthritis (RA). Methods Fifty RA patients and ten healthy volunteers were selected according to inclusion criteria. Peripheral blood macrophages were extracted and divided into six groups: normal control(NC), RA macrophage model (RA-MC), RA-MC with NLRC3 overexpression, RA-MC with NLRC3 knockdown, RA-MC with STING overexpression, and RA-MC with STING knockdown groups. Macrophage pyroptosis was observed using transmission electron microscopy. The mRNA expressions of NLRC3, STING, caspase-1, and GSDMD were detected using RT-qPCR. interleukin 1ß (IL-1ß) and IL-18 levels in cell supernatants were measured using ELISA. Results Compared to the NC group, the RA-MC group showed characteristics of pyroptosis. Compared to the RA-MC group, the groups of RA-MC with NLRC3 overexpression and RA-MC with STING knockdown showed improved pyroptosis, while the groups of RA-MC with NLRC3 knockdown and RA-MC with STING overexpression demonstrated exacerbated pyroptosis. Compared to the NC group, the RA-MC group showed increased mRNA expression levels of STING, caspase-1 and GSDMD, as well as increased levels of the inflammatory cytokines IL-1ß and IL-18, but decreased NLRC3 mRNA expression level. Compared to the RA-MC group, the groups of RA-MC with NLRC3 overexpression and RA-MC with STING knockdown showed reduced mRNA expression levels of caspase-1 and GSDMD, as well as reduced inflammatory factors, while the groups of RA-MC with NLRC3 knockdown and RA-MC with STING overexpression had opposite results. Compared to the RA-MC group, the group of RA-MC with NLRC3 overexpression showed a decreased STING mRNA expression level, while the group of RA-MC with NLRC3 knockdown demonstrated an increased level. Conclusion NLRC3 can inhibit the STING signaling pathway, reduce pyroptosis proteins caspase-1 and GSDMD, antagonize macrophage pyroptosis, and lower the levels of inflammatory cytokines IL-1ß and IL-18, thereby alleviating the immune inflammatory response in RA patients.
Asunto(s)
Artritis Reumatoide , Interleucina-1beta , Macrófagos , Proteínas de la Membrana , Piroptosis , Transducción de Señal , Humanos , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Piroptosis/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Femenino , Masculino , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Persona de Mediana Edad , Interleucina-18/genética , Interleucina-18/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Adulto , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Gasderminas , Péptidos y Proteínas de Señalización IntercelularRESUMEN
Pyroptosis is a gasdermin-mediated pro-inflammatory form of programmed cell death (PCD). Tumor necrosis factor-É (TNF-É) is an inflammatory cytokine, and some studies have shown that TNF-É can cause pyroptosis of cells and exert anti-tumor effects. However, whether TNF-É exerts anti-tumor effects on breast cancer cells by inducing pyroptosis has not been reported. In this study, to explore the impact of TNF-É on pyroptosis in breast cancer cells, we treated MCF-7 cells with TNF-É and found that TNF-É induced cell death. Moreover, we observed that the dead cells were swollen with obvious balloon-like bubbles, which was a typical sign of pyroptosis. Further studies have found that the anti-tumor effect of TNF-É on breast cancer cells in vitro was achieved through the canonical pyroptosis pathway. In addition, TNF-É-induced pyroptosis in MCF-7 cells was associated with mitochondrial dysfunction, in which mitochondrial membrane potential was decreased and mitochondrial ROS production was increased. After inhibiting ROS production, the activation effect of TNF-É on NLRP3/Caspase-1/GSDMD pathway was weakened, and the inhibitory effect of TNF-É on the growth of MCF-7 cells in vitro was also decreased, further confirming the involvement of ROS in TNF-É-induced pyroptosis. Overall, our study revealed a new mechanism by which TNF-É exerts an anti-tumor effect by inducing pyroptosis in MCF-7 cells through the ROS/NLRP3/Caspase-1/GSDMD pathway, which may provide new therapeutic ideas for the treatment of breast cancer.
Asunto(s)
Caspasa 1 , Péptidos y Proteínas de Señalización Intracelular , Mitocondrias , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas de Unión a Fosfato , Piroptosis , Especies Reactivas de Oxígeno , Factor de Necrosis Tumoral alfa , Femenino , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Caspasa 1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Piroptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismoRESUMEN
This study reports the facile hydrothermal synthesis of pure Bi2WO6 and Bi2WO6\MWCNTs nanocomposite at specific molar ratio 1:2.5 of Bi2WO6:MWCNTs and elucidates their role in modulating the NLRP3 inflammasome pathway via autophagy induction. Comprehensive characterization techniques, including XRD, Raman, UV.Vis PL,FESEM,EDS and TEM, revealed the successful incorporation of MWCNTs into the Bi2WO6 structures, leading to enhanced crystattlinity, reduced band gap energy (2.4 eV) suppressed charge carrier recombination and mitigated nanoparticles aggregation. Notably, the reduced band gap facikitaed improved visible light harvesting, a crucial attribute for photocatalytic applications. Significantly, the nanocompsoite exhibited a remarkable capacity to augment autophagy in bone marrow-derived macrophages (BMDMs), consequently down-regulating the NLRP3 inflammasom activation and IL-1ß secretion upon LPS and ATP stimulation. Immunofluorescence assays unveiled increased co-localization of LC3 and NLRP3, suggestion enhanced targeting of NLRP3 by autophagy. Inhibition of autophagy by 3-MA reversed these effects, confirming the pivotal role of autophagy induction. Furthermore, the nanocomposite attenuated caspase-1 activation and ASC oligomerzation, thereby impeding inflammasome assembly. Collectively, these findings underscore the potential of Bi2WO6\MWCNTs nanocompsite as a multifaceted therapeutic platform, levering its tailored optoelectronic properties and sbility to modulate the NLRP3 infalmmasome via autophagy augmentation. This work covers the way for the development of advanced nanomaterials with tunable functionalities for combating inflammatory disorders and antimicrobial applications.
Asunto(s)
Autofagia , Inflamasomas , Interleucina-1beta , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Autofagia/efectos de los fármacos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Animales , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Nanopartículas/química , Compuestos de Tungsteno/farmacología , Compuestos de Tungsteno/química , Caspasa 1/metabolismo , Nanocompuestos/químicaRESUMEN
INTRODUCTION: The presence of hypoxic-ischemic brain damage (HIBD) in neonates triggers a strong neuroinflammatory reaction. Pyroptosis, a programmed cell death mechanism associated with inflammation, plays a crucial role in HIBD. Pyruvate kinase M2 (PKM2) plays a significant role in connecting metabolic processes and inflammatory responses, but whether it affects hippocampus pyroptosis in HIBD is unclear. The aim of this study is to elucidate the role of PKM2 in HIBD and to propose a novel therapeutic approach for neonatal ischemic-hypoxic encephalopathy. METHODS: In this study, we employed neonatal 7-day-old Sprague Dawley rats to establish a model of HIBD using the Rice-Vannucci surgical technique and a hypoxia device. To inhibit the elevation of PKM2, we utilized the PKM2 inhibitor shikonin. The rats were categorized into four groups: Sham, Shikonin, HIBD, and Shikonin + HIBD. Behavioral tests, hematoxylin eosin staining, immunofluorescence staining, ELISA (IL-1ß, IL-18), and LDH were conducted in each group to evaluate neurological function, hippocampal damage, the occurrence of neuronal pyroptosis, and the neuroinflammation. Western blot was used to assess the expression levels of PKM2, NLRP3, Caspase-1, Cleaved Caspase-1, GSDMD, GSDMDN, and IL-1ß. RESULTS: The expression of PKM2 elevated in hippocampal tissues of the HIBD model and the localization of PKM2 in the hippocampus was activated in neurons instead of microglia during the HIBD. Meanwhile, the inhibition of PKM2 improved the behavioral test scores and the body weight of rats, the neuronal damage in the CA1 region of hippocampal tissue was also attenuated. In addition, inhibiting PKM2 alleviated neuronal pyroptosis by decreasing the expression of PKM2, NLRP3, Caspase-1, Cleaved Caspase-1, GSDMD, GSDMDN. Furthermore, serum levels of LDH and inflammatory factors IL-1ß and IL-18 decrease with PKM2 inhibition. CONCLUSIONS: Based on these findings, we can conclude that PKM2 plays a crucial role in regulating hippocampal neuronal pyroptosis of HIBD rats via NLRP3/Caspase-1/GSDMD pathway. Therefore, inhibiting PKM2 could be a promising therapeutic strategy for the treatment of neonatal ischemic-hypoxic encephalopathy.
Asunto(s)
Animales Recién Nacidos , Caspasa 1 , Hipocampo , Hipoxia-Isquemia Encefálica , Proteína con Dominio Pirina 3 de la Familia NLR , Neuronas , Piroptosis , Piruvato Quinasa , Ratas Sprague-Dawley , Animales , Piroptosis/efectos de los fármacos , Piroptosis/fisiología , Hipoxia-Isquemia Encefálica/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas , Hipocampo/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Caspasa 1/metabolismo , Piruvato Quinasa/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Naftoquinonas/farmacología , Naftoquinonas/administración & dosificación , GasderminasRESUMEN
The objective of this study is to elucidate how Royal jelly (RJ) and 10-hydroxy-2-decanoic acid (10-HDA) prevents diabetic skin dysfunction by modulating the pyroptosis pathway. Type 2 diabetes models are induced by fat diet consumption and low dose of streptozotocin (STZ) in C57BL/6J mice and treated with RJ (100 mg kg-1 day-1) and 10-HDA, the major lipid component of royal jelly (100 mg kg-1 day-1) for 28 weeks. The results show that serum concentrations of glucose and triglyceride are significantly lower in the RJ group or 10-HDA than diabetes mellitus (DM) group. Compared to the control group, pyroptosis proteins, GSDMD, ASC, Caspase-1, and IL-1ß are increased in the skin of the diabetic model, accompanied by the activation of the Wnt/ß-catenin signal pathway. Further evaluations by RJ exhibit superior improvement of skin damage, repress activation of the Wnt/ß-catenin pathway, and attenuate keratinocyte pyroptosis, but 10-HDA cannot completely suppress the activation of Wnt/ß-catenin pathway and pyroptosis, which shows a relatively weak protective effect on skin damage which shows that RJ is a better effect on skin injury after DM.
Asunto(s)
Diabetes Mellitus Experimental , Ácidos Grasos , Queratinocitos , Ratones Endogámicos C57BL , Piroptosis , Piel , Vía de Señalización Wnt , Animales , Piroptosis/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Ácidos Grasos/farmacología , Queratinocitos/efectos de los fármacos , Masculino , Piel/efectos de los fármacos , Piel/metabolismo , Interleucina-1beta/metabolismo , Caspasa 1/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Ratones , Diabetes Mellitus Tipo 2/metabolismo , beta Catenina/metabolismo , Glucemia/efectos de los fármacos , Triglicéridos/sangre , Triglicéridos/metabolismo , Gasderminas , Ácidos Grasos MonoinsaturadosRESUMEN
Inflammasome activation results in the cleavage of gasdermin D (GSDMD) by pro-inflammatory caspases. The N-terminal domains (GSDMDNT) oligomerize and assemble pores penetrating the target membrane. As methods to study pore formation in living cells are insufficient, the order of conformational changes, oligomerization, and membrane insertion remained unclear. We have raised nanobodies (VHHs) against human GSDMD and find that cytosolic expression of VHHGSDMD-1 and VHHGSDMD-2 prevents oligomerization of GSDMDNT and pyroptosis. The nanobody-stabilized GSDMDNT monomers partition into the plasma membrane, suggesting that membrane insertion precedes oligomerization. Inhibition of GSDMD pore formation switches cell death from pyroptosis to apoptosis, likely driven by the enhanced caspase-1 activity required to activate caspase-3. Recombinant antagonistic nanobodies added to the extracellular space prevent pyroptosis and exhibit unexpected therapeutic potential. They may thus be suitable to treat the ever-growing list of diseases caused by activation of (non-) canonical inflammasomes.
Asunto(s)
Inflamasomas , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Unión a Fosfato , Piroptosis , Anticuerpos de Dominio Único , Humanos , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/química , Inflamasomas/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Piroptosis/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Células HEK293 , Caspasa 1/metabolismo , Caspasa 3/metabolismo , Membrana Celular/metabolismo , Multimerización de Proteína , Apoptosis/efectos de los fármacos , GasderminasRESUMEN
Cleavage of the innate immune receptor NLRP1B by various microbial proteases causes the proteasomal degradation of its N-terminal fragment and the subsequent release of a C-terminal fragment that forms an inflammasome. We reported previously that metabolic stress caused by intracellular bacteria triggers NLRP1B activation, but the mechanism by which this occurs was not elucidated. Here we demonstrate that TLR4 signaling in metabolically stressed macrophages promotes the formation of a TRIF/RIPK1/caspase-8 complex. Caspase-8 activity, induced downstream of this TLR4 pathway or through a distinct TNF receptor pathway, causes cleavage and activation of NLRP1B, which facilitates the maturation of both pro-caspase-1 and pro-caspase-8. Thus, our findings indicate that caspase-8 and NLRP1B generate a positive feedback loop that amplifies cell death processes and promotes a pro-inflammatory response through caspase-1. The ability of NLRP1B to detect caspase-8 activity suggests that this pattern recognition receptor may play a role in the defense against a variety of pathogens that induce apoptosis.
Asunto(s)
Proteínas Reguladoras de la Apoptosis , Caspasa 8 , Inflamasomas , Macrófagos , Caspasa 8/metabolismo , Caspasa 8/genética , Inflamasomas/metabolismo , Animales , Ratones , Macrófagos/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Transducción de Señal , Ratones Endogámicos C57BL , Receptor Toll-Like 4/metabolismo , Caspasa 1/metabolismo , Humanos , Ratones Noqueados , ApoptosisRESUMEN
No single treatment significantly reduces the mortality rate and improves neurological outcomes after intracerebral haemorrhage (ICH). New evidence suggests that pyroptosis-specific proteins are highly expressed in the perihaematomal tissues of patients with ICH and that the disulfiram (DSF) inhibits pyroptosis. An ICH model was established in C57BL/6 mice by intracranial injection of collagenase, after which DSF was used to treat the mice. Cell model of ICH was constructed, and DSF was used to treat the cells. HE, TUNEL, Nissl, FJC and IF staining were performed to evaluate the morphology of brain tissues; Western blotting and ELISA were performed to measure the protein expression of NOD-like receptor protein 3 (NLRP3)/Caspase-1/gasdermin D (GSDMD) classical pyroptosis pathway and Toll-likereceptor4 (TLR4)/nuclear factor-kappaB (NF-κB) inflammatory signaling pathway and bloodâbrain barrier-associated factoes, and the wet/dry weight method was used to determine the brain water content. The expression of proteins related to the NLRP3/Caspase-1/GSDMD pathway and the TLR4/NF-κB pathway was upregulated in tissues surrounding the haematoma compared with that in control tissues; Moreover, the expression of the blood-brain barrier structural proteins occludin and zonula occludens-1 (ZO-1) was downregulated, and the expression of Aquaporin Protein-4 (AQP4) and matrix metalloprotein 9 (MMP-9) was upregulated. DSF significantly inhibited these changes, reduced the haematoma volume, decreased the brain water content, reduced neuronal death and degeneration and improved neurological function after ICH. ICH activated the classical pyroptosis pathway and TLR4/NF-κB inflammatory pathway, disruped the expression of blood-brain barrier structural proteins, and exacerbated brain injury and neurological dysfunction. DSF inhibited these changes and exerted the therapeutic effects on pathological changes and dysfunction caused by ICH.
Asunto(s)
Barrera Hematoencefálica , Disulfiram , Ratones Endogámicos C57BL , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Transducción de Señal , Receptor Toll-Like 4 , Animales , Piroptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Disulfiram/farmacología , Transducción de Señal/efectos de los fármacos , Masculino , Receptor Toll-Like 4/metabolismo , FN-kappa B/metabolismo , Modelos Animales de Enfermedad , Caspasa 1/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/metabolismo , Ocludina/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Humanos , GasderminasRESUMEN
BACKGROUND: The novel phthalein component QBT, extracted from Ligusticum chuanxiong, shows promising biological activity against cerebrovascular diseases. This study focused on ferroptosis and pyroptosis to explore the effects of QBT on nerve injury, cognitive dysfunction, and related mechanisms in a rat model of vascular dementia (VaD). METHODS: We established a rat model of VaD and administered QBT as a treatment. Cognitive dysfunction in VaD rats was evaluated using novel object recognition and Morris water maze tests. Neuronal damage and loss in the brain tissues of VaD rats were assessed with Nissl staining and immunofluorescence. Furthermore, we investigated the neuroprotective mechanisms of QBT by modulating the nuclear factor erythroid 2-related factor 2 (Nrf2)/cystine-glutamate antiporter (xCT)/glutathione peroxidase 4 (GPX4) and Nod-like receptor family pyrin domain-containing 3 (NLRP3)/cysteine-requiring aspartate protease-1 (Caspase-1)/Gasdermin D (GSDMD) pathways to inhibit ferroptosis and pyroptosis both in vivo and in vitro. RESULTS: Our findings indicated that QBT significantly ameliorated neuronal damage and cognitive dysfunction in VaD rats. Additionally, QBT reversed abnormal changes associated with ferroptosis and pyroptosis in the brains of VaD rats, concurrently up-regulating the Nrf2/xCT/GPX4 pathway and down-regulating the NLRP3/Caspase-1/GSDMD pathway to inhibit ferroptosis and pyroptosis in neuronal cells, thereby exerting a neuroprotective role. CONCLUSION: In summary, QBT effectively mitigated neuronal damage and cognitive dysfunction in VaD rats, demonstrating a neuroprotective effect by inhibiting ferroptosis and pyroptosis in neuronal cells. This study offers a novel perspective and theoretical foundation for the future development of drugs targeting VaD.
Asunto(s)
Caspasa 1 , Disfunción Cognitiva , Demencia Vascular , Ferroptosis , Factor 2 Relacionado con NF-E2 , Proteína con Dominio Pirina 3 de la Familia NLR , Neuronas , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Piroptosis , Ratas Sprague-Dawley , Transducción de Señal , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Ferroptosis/efectos de los fármacos , Demencia Vascular/tratamiento farmacológico , Demencia Vascular/metabolismo , Demencia Vascular/psicología , Piroptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/metabolismo , Masculino , Ratas , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Caspasa 1/metabolismo , Transducción de Señal/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/farmacología , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Humanos , Gasderminas , Proteínas de Unión a FosfatoRESUMEN
Zymosan is a ß-glucan-rich component derived from the cell walls of Saccharomyces cerevisiae extensively used in research for its potent immunomodulatory properties. It can prompt inflammatory responses such as peritonitis and arthritis, and is particularly used to study the immune response to fungal particles. Although the zymosan induced-release of the proinflammatory cytokine IL-1ß by macrophages is an essential mechanism for combating fungal infection and inducing inflammation, the exact processes leading to its release remain not well understood. In this study, we uncover the intracellular mechanisms involved in zymosan induced-release of active IL-1ß by peritoneal macrophages. Zymosan initiates pro-IL-1ß formation through TLR2/MyD88 activation; however, Dectin-1 activation only amplify the conversion of pro-IL-1ß into its active form. The conversion of inactive to active IL-1ß upon zymosan stimulation depends on the NLRP3, ASC, and caspase-1 driven by the decrease in intracellular potassium ions. Notably, zymosan-induced activation of caspase-1 does not require phagocytosis. Instead, zymosan induces a rapid drop in the intracellular ATP concentration, which occurs concomitant with caspase-1 and IL-1ß activation. Accordingly, disruption of glycolytic flux during zymosan stimulation promotes an additional reduction of intracellular ATP and concurrently amplifies the activation of caspase-1 and IL-1ß. These results reveal that fungal recognition by macrophages results in a metabolic dysfunction, leading to a decrease of intracellular ATP associated with inflammasome activation.
Asunto(s)
Adenosina Trifosfato , Proteínas Adaptadoras de Señalización CARD , Caspasa 1 , Inflamasomas , Interleucina-1beta , Lectinas Tipo C , Proteína con Dominio Pirina 3 de la Familia NLR , Zimosan , Inflamasomas/metabolismo , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-1beta/metabolismo , Caspasa 1/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Ratones , Lectinas Tipo C/metabolismo , Adenosina Trifosfato/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/metabolismo , Ratones Endogámicos C57BL , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Glucólisis/efectos de los fármacos , Fagocitosis/efectos de los fármacosRESUMEN
To research the role of the NLRP3 inflammasome in Schistosoma japonicum-induced granuloma formation and liver fibrosis. In in vivo tests, BALB/c mice were used. shNLRP3 plasmid based on adeno-associated virus serotype 8 (AAV8-shNLRP3) was injected to block NLRP3 inflammasome via tail vein. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were detected to assess liver injury. H&E staining was used for routine histopathological assessment; Masson's trichrome staining was used to detect fibrous tissues and collagen fibers. Hepatic expression of NLRP3, procaspase-1, bioactive caspase-1, collagen-1, tissue inhibitor of metalloproteinases-1 (TIMP-1), and α-smooth muscle actin (α-SMA) were detected by western blot. Serum levels of IL-1ß were detected by enzyme-linked immunosorbent assay (ELISA). The inflammatory cell infiltration and hepatic expression of IL-1ß around the granuloma were detected by immunohistochemistry staining. Treatment of S. japonicum infected mice with AAV8-shNLRP3 significantly reduced the hepatic levels of bioactive caspase-1 and IL-1ß, as well as circulating IL-1ß concentrations, while reducing the amounts of myeloperoxidase (MPO) and F4/80 positive cells around the granuloma. Moreover, collagen deposition, TIMP-1, and α-SMA, which are markers of hepatic stellate cell (HSC) activation, were reduced around the liver granuloma. These findings highlight a therapeutic potential of AAV8-shNLRP3 in schistosomiasis cirrhosis.
Asunto(s)
Caspasa 1 , Inflamasomas , Cirrosis Hepática , Ratones Endogámicos BALB C , Proteína con Dominio Pirina 3 de la Familia NLR , Schistosoma japonicum , Esquistosomiasis Japónica , Inhibidor Tisular de Metaloproteinasa-1 , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Esquistosomiasis Japónica/patología , Esquistosomiasis Japónica/inmunología , Inflamasomas/metabolismo , Ratones , Caspasa 1/metabolismo , Caspasa 1/genética , Cirrosis Hepática/patología , Cirrosis Hepática/parasitología , Cirrosis Hepática/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Granuloma/patología , Hígado/patología , Hígado/parasitología , Hígado/metabolismo , Inflamación , Femenino , Actinas/metabolismo , Modelos Animales de EnfermedadRESUMEN
OBJECTIVE: The purpose of the present study was to potential effects of forsythiaside A (FA) on Sjogren's syndrome (SS). METHODS: Enzyme linked immunosorbent assay for detecting cytokines and Western blotting was used for detecting related protein expression. RESULTS: FA effectively reduced the secretion of inflammatory cytokines, the expression of Caspase-1 and NLRP3 proteins and the expression of p65 in SS. FA also effectively inhibited the high expression of Grp78 in SS. When Grp78 expression was silenced, it effectively reduced the secretion of inflammatory cytokines, the expression of Caspase-1 and NLRP3 proteins and the expression of p65 in the nucleus in SS. FA effectively inhibit the secretion of inflammatory cytokines induced by overexpression of Grp78, the expression of Caspase-1 and NLRP3 proteins and the expression of p65 in the nucleus in SS. CONCLUSION: FA induces the degradation of Grp78 protein, regulates the NF-κB signaling pathway in SS and inhibited NLRP3 inflammasome activation and reduced the release of inflammatory cytokines to alleviate SS.
Asunto(s)
Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Síndrome de Sjögren , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Humanos , Chaperón BiP del Retículo Endoplásmico/metabolismo , Inflamasomas/metabolismo , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Femenino , Transducción de Señal , Citocinas/metabolismo , Caspasa 1/metabolismo , Persona de Mediana Edad , Masculino , Factor de Transcripción ReIA/metabolismo , FN-kappa B/metabolismoRESUMEN
The NLRP3 inflammasome is dysregulated in autoinflammatory disorders caused by inherited mutations and contributes to the pathogenesis of several chronic inflammatory diseases. In this study, we discovered that disulfiram, a safe US Food and Drug Administration (FDA)-approved drug, specifically inhibits the NLRP3 inflammasome but not the NLRC4 or AIM2 inflammasomes. Disulfiram suppresses caspase-1 activation, ASC speck formation, and pyroptosis induced by several stimuli that activate NLRP3. Mechanistically, NLRP3 is palmitoylated at cysteine 126, a modification required for its localization to the trans-Golgi network and inflammasome activation, which was inhibited by disulfiram. Administration of disulfiram to animals inhibited the NLRP3, but not NLRC4, inflammasome in vivo. Our study uncovers a mechanism by which disulfiram targets NLRP3 and provides a rationale for using a safe FDA-approved drug for the treatment of NLRP3-associated inflammatory diseases.