Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 497
Filtrar
1.
ACS Nano ; 18(19): 12412-12426, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38693619

RESUMEN

Glycans play vital roles in nearly all life processes of multicellular organisms, and understanding these activities is inseparable from elucidating the biological significance of glycans. However, glycan research has lagged behind that of DNA and protein due to the challenges posed by structural heterogeneity and isomerism (i.e., structures with equal molecular weights) the lack of high-efficiency structural analysis techniques. Nanopore technology has emerged as a sensitive single-molecule biosensor, shining a light on glycan analysis. However, a significant number of glycans are small and uncharged, making it challenging to elicit identifiable nanopore signals. Here we introduce a R-binaphthyl tag into glycans, which enhances the cation-π interaction between the derivatized glycan molecules and the nanopore interface, enabling the detection of neutral glycans with an aerolysin nanopore. This approach allows for the distinction of di-, tri-, and tetrasaccharides with monosaccharide resolution and has the potential for group discrimination, the monitoring of enzymatic transglycosylation reactions. Notably, the aerolysin mutant T240R achieves unambiguous identification of six disaccharide isomers, trisaccharide and tetrasaccharide linkage isomers. Molecular docking simulations reveal that multiple noncovalent interactions occur between residues R282, K238, and R240 and the glycans and R-binaphthyl tag, significantly slowing down their translocation across the nanopore. Importantly, we provide a demonstration of the kinetic translocation process of neutral glycan isomers, establishing a solid theoretical foundation for glycan nanopore analysis. The development of our technology could promote the analysis of glycan structural isomers and has the potential for nanopore-based glycan structural determination and sequencing.


Asunto(s)
Toxinas Bacterianas , Nanoporos , Polisacáridos , Proteínas Citotóxicas Formadoras de Poros , Polisacáridos/química , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Simulación del Acoplamiento Molecular , Mutación
2.
In Vivo ; 38(3): 1042-1048, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38688646

RESUMEN

BACKGROUND/AIM: Oral epithelial cells serve as the primary defense against microbial exposure in the oral cavity, including the fungus Candida albicans. Dectin-1 is crucial for recognition of ß-glucan in fungi. However, expression and function of Dectin-1 in oral epithelial cells remain unclear. MATERIALS AND METHODS: We assessed Dectin-1 expression in Ca9-22 (gingiva), HSC-2 (mouth), HSC-3 (tongue), and HSC-4 (tongue) human oral epithelial cells using flow cytometry and real-time polymerase chain reaction. Cell treated with ß-glucan-rich zymosan were evaluated using real-time polymerase chain reaction. Phosphorylation of spleen-associated tyrosine kinase (SYK) was analyzed by western blotting. RESULTS: Dectin-1 was expressed in all four cell types, with high expression in Ca9-22 and HSC-2. In Ca9-22 cells, exposure to ß-glucan-rich zymosan did not alter the mRNA expression of chemokines nor of interleukin (IL)6, IL8, IL1ß, IL17A, and IL17F. Zymosan induced the expression of antimicrobial peptides ß-defensin-1 and LL-37, but not S100 calcium-binding protein A8 (S100A8) and S100A9. Furthermore, the expression of cylindromatosis (CYLD), a negative regulator of nuclear factor kappa B (NF-κB) signaling, was induced. In HSC-2 cells, zymosan induced the expression of IL17A. The expression of tumor necrosis factor alpha-induced protein 3 (TNFAIP3), a negative regulator of NF-κB signaling, was also induced. Expression of other cytokines and antimicrobial peptides remained unchanged. Zymosan induced phosphorylation of SYK in Ca9-22 cells, as well as NF-κB. CONCLUSION: Oral epithelial cells express Dectin-1 and recognize ß-glucan, which activates SYK and induces the expression of antimicrobial peptides and negative regulators of NF-κB, potentially maintaining oral homeostasis.


Asunto(s)
Células Epiteliales , Lectinas Tipo C , FN-kappa B , Transducción de Señal , Quinasa Syk , Humanos , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , FN-kappa B/metabolismo , Quinasa Syk/metabolismo , Quinasa Syk/genética , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Línea Celular , Zimosan/farmacología , Citocinas/metabolismo , Citocinas/genética , Fosforilación , Mucosa Bucal/metabolismo , Mucosa Bucal/inmunología , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo
3.
Front Immunol ; 15: 1365975, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38404589

RESUMEN

Germline mutations in genes involved in perforin-granzyme-mediated cytotoxicity such as PRF1, UNC13D, STX11, and STXBP2 were known to cause familial hemophagocytic lymphohistiocytosis (FHL). In this study, we reported a unique group of 3 patients with germline mutations of UNC13D and STX11 genes and presented as adult-onset peripheral T-cell lymphoma (PTCL) with cytotoxic T-cell phenotype and atypical lymphoma presentations. CD107a degranulation assay and NK-cell activity analysis demonstrated impaired cytotoxic function of the NK/T-cells of the patients with FHL-related mutations. Gene expression profile study revealed that up-regulated genes of the cytotoxic T-cells were enriched in autoimmune-related pathways. It was possible that impaired cytotoxic lymphocyte-mediated immune surveillance and autoantigen stimulation may both participate in PTCL oncogenesis. Germline defects of FLH-related genes may represent a novel predisposing factor for PTCLs.


Asunto(s)
Linfohistiocitosis Hemofagocítica , Linfoma de Células T Periférico , Adulto , Humanos , Proteínas Citotóxicas Formadoras de Poros/genética , Células Asesinas Naturales , Células Germinativas/metabolismo , Proteínas de la Membrana
4.
Nat Commun ; 15(1): 1405, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38360940

RESUMEN

Mechanical force contributes to perforin pore formation at immune synapses, thus facilitating the cytotoxic T lymphocytes (CTL)-mediated killing of tumor cells in a unidirectional fashion. How such mechanical cues affect CTL evasion of perforin-mediated autolysis remains unclear. Here we show that activated CTLs use their softness to evade perforin-mediated autolysis, which, however, is shared by T leukemic cells to evade CTL killing. Downregulation of filamin A is identified to induce softness via ZAP70-mediated YAP Y357 phosphorylation and activation. Despite the requirements of YAP in both cell types for softness induction, CTLs are more resistant to YAP inhibitors than malignant T cells, potentially due to the higher expression of the drug-resistant transporter, MDR1, in CTLs. As a result, moderate inhibition of YAP stiffens malignant T cells but spares CTLs, thus allowing CTLs to cytolyze malignant cells without autolysis. Our findings thus hint a mechanical force-based immunotherapeutic strategy against T cell leukemia.


Asunto(s)
Citotoxicidad Inmunológica , Linfocitos T Citotóxicos , Perforina/genética , Perforina/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética
5.
Gene ; 893: 147888, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-37839766

RESUMEN

BACKGROUND: Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), acting as one common sepsis-associated organ injury, induces uncontrolled and self-amplifies pulmonary inflammation. Given the lack of clinically effective approaches, the mortality rate of it still remains high. Suramin(SUR), as an antiparasitic drug initially, was found to ameliorate sepsis associated ALI in our previous work. However, the underlying mechanism of its protective effects has not been clarified. Pyroptosis, categorized as an inflammatory form of programmed cell death, could aggravate lung inflammatory responses via inducing alveolar macrophages (AM) pyroptosis. METHODS: MH-S AM cell line was stimulated with or without lipopolysaccharide (LPS) or suramin, and the differential expression genes (DEGs) were excavated using RNA sequencing (RNA-seq). To identify the regulatory roles of these genes, pyroptosis-related genes (PRGs), GO/KEGG and GSEA analysis were conducted. We also performed WB, qRTPCR and ELISA to validate the RNA-seq results and further expound the protective effect of suramin. RESULTS: 624 DEGs were identified between control (CON) and lipopolysaccharide (LPS) groups, and enrichment analysis of these genes revealed significantly enriched pathways that related to immune system and signal transduction. Meanwhile, 500 DEGs were identified in LPS/SUR+LPS group. In addition to the pathways mentioned above, IL-17 pathway and C-type lectin receptor signaling pathway were also enriched. All 6 pathways were connected with pyroptosis. Concurrently, the "DESeq2" R package was used to identify differentially expressed PRGs. Nod1, Nod2, interleukin (IL)-1b, IL-6, tumor necrosis factor (TNF), NLRP3 were upregulated under LPS stimulation. Then, in SUR+LPS group, Nod2, IL-6, IL-1b, NLRP3 were downregulated. The validation results of WB, qRT-PCR, and ELISA showed: the protein and mRNA expression levels of NLRP3, caspase-1, GSDMD and the concentrations of IL-1b, IL-18 were decreased when treated with suramin and LPS. CONCLUSION: Suramin could inhibit NLRP3/caspase-1/GSDMD canonical pyroptosis pathway in LPS-induced MH-S alveolar macrophages.


Asunto(s)
Macrófagos Alveolares , Sepsis , Humanos , Macrófagos Alveolares/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Lipopolisacáridos/farmacología , Suramina/farmacología , Interleucina-6/genética , RNA-Seq , Inflamasomas/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/farmacología , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/farmacología
6.
Cell Biol Toxicol ; 39(6): 2743-2760, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37462807

RESUMEN

Gasdermin (GSDM) family, the key executioners of pyroptosis, play crucial roles in anti-pathogen and anti-tumor immunities, although little is known about the expression of GSDM in lung diseases at single-cell resolution, especially in lung epithelial cells. We comprehensively investigated the transcriptomic profiles of GSDM members in various lung tissues from healthy subjects or patients with different lung diseases at single cell level, e.g., chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), lung adenocarcinoma (LUAD), or systemic sclerosis (SSC). The expression of GSDM members varied among pulmonary cell types (immune cells, structural cells, and especially epithelial cells) and even across lung diseases. Regarding disease-associated specificities, we found that GSDMC or GSDMD altered significantly in ciliated epithelia of COPD or LUAD, GSDMD in mucous, club, and basal cells of LUAD and GSDMC in mucous epithelia of para-tumor tissue, as compared with the corresponding epithelia of other diseases. The phenomic specificity of GSDM in lung cancer subtypes was noticed by comparing with 15 non-pulmonary cancers and para-cancer samples. GSDM family gene expression changes were also observed in different lung epithelial cell lines (e.g., HBE, A549, H1299, SPC-1, or H460) in responses to external challenges, including lipopolysaccharide (LPS), lysophosphatidylcholine (lysoPC), cigarette smoking extract (CSE), cholesterol, and AR2 inhibitor at various doses or durations. GSDMA is rarely expressed in those cell lines, while GSDMB and GSDMC are significantly upregulated in human lung epithelia. Our data indicated that the heterogeneity of GSDM member expression exists at different cells, pathologic conditions, challenges, probably dependent upon cell biological phenomes, functions, and behaviors, upon cellular responses to external changes, and the nature and severity of lung disease. Thus, the deep exploration of GSDM phenomes may provide new insights into understanding the single-cell roles in the tissue, regulatory roles of the GSDM family in the pathogenesis, and potential values of biomarker identification and development.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Proteínas de Neoplasias/metabolismo , Transcriptoma/genética , Células Epiteliales/metabolismo , Neoplasias Pulmonares/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Biomarcadores de Tumor/genética , Proteínas Citotóxicas Formadoras de Poros/genética
7.
Science ; 380(6651): 1258-1265, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37347855

RESUMEN

During initiation of antiviral and antitumor T cell-mediated immune responses, dendritic cells (DCs) cross-present exogenous antigens on major histocompatibility complex (MHC) class I molecules. Cross-presentation relies on the unusual "leakiness" of endocytic compartments in DCs, whereby internalized proteins escape into the cytosol for proteasome-mediated generation of MHC I-binding peptides. Given that type 1 conventional DCs excel at cross-presentation, we searched for cell type-specific effectors of endocytic escape. We devised an assay suitable for genetic screening and identified a pore-forming protein, perforin-2 (Mpeg1), as a dedicated effector exclusive to cross-presenting cells. Perforin-2 was recruited to antigen-containing compartments, where it underwent maturation, releasing its pore-forming domain. Mpeg1-/- mice failed to efficiently prime CD8+ T cells to cell-associated antigens, revealing an important role for perforin-2 in cytosolic entry of antigens during cross-presentation.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos , Endocitosis , Proteínas Citotóxicas Formadoras de Poros , Animales , Ratones , Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Endocitosis/genética , Endocitosis/inmunología , Pruebas Genéticas , Antígenos de Histocompatibilidad Clase I , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteolisis
8.
J Infect Dis ; 228(8): 990-998, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36967681

RESUMEN

BACKGROUND: Genome-wide association studies have identified several risk alleles for early childhood asthma, particularly in the 17q21 locus and in the cadherin-related family member 3 (CDHR3) gene. Contribution of these alleles to the risk of acute respiratory tract infections (ARI) in early childhood is unclear. METHODS: We analyzed data from the STEPS birth-cohort study of unselected children and the VINKU and VINKU2 studies on children with severe wheezing illness. Genome-wide genotyping was performed on 1011 children. We analyzed the association between 11 preselected asthma risk alleles and the risk of ARIs and wheezing illnesses of various viral etiologies. RESULTS: The asthma risk alleles in CDHR3, GSDMA, and GSDMB were associated with an increased rate of ARIs (for CDHR3, incidence rate ratio [IRR], 1.06; 95% confidence interval [CI], 1.01-1.12; P = .02), and risk allele in CDHR3 gene with rhinovirus infections (IRR, 1.10; 95% CI, 1.01-1.20, P = .03). Asthma risk alleles in GSDMA, GSDMB, IKZF3, ZPBP2, and ORMDL3 genes were associated with wheezing illnesses in early childhood, especially rhinovirus-positive wheezing illnesses. CONCLUSIONS: Asthma risk alleles were associated with an increased rate of ARIs and an increased risk of viral wheezing illnesses. Nonwheezing and wheezing ARIs and asthma may have shared genetic risk factors. Clinical Trials Registration. NCT00494624 and NCT00731575.


Asunto(s)
Asma , Infecciones del Sistema Respiratorio , Humanos , Niño , Preescolar , Alelos , Estudios de Cohortes , Ruidos Respiratorios/genética , Estudio de Asociación del Genoma Completo , Asma/epidemiología , Asma/genética , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/genética , Proteínas del Huevo/genética , Proteínas de la Membrana/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Relacionadas con las Cadherinas
9.
Nature ; 616(7957): 598-605, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36991125

RESUMEN

Cytotoxic lymphocyte-derived granzyme A (GZMA) cleaves GSDMB, a gasdermin-family pore-forming protein1,2, to trigger target cell pyroptosis3. GSDMB and the charter gasdermin family member GSDMD4,5 have been inconsistently reported to be degraded by the Shigella flexneri ubiquitin-ligase virulence factor IpaH7.8 (refs. 6,7). Whether and how IpaH7.8 targets both gasdermins is undefined, and the pyroptosis function of GSDMB has even been questioned recently6,8. Here we report the crystal structure of the IpaH7.8-GSDMB complex, which shows how IpaH7.8 recognizes the GSDMB pore-forming domain. We clarify that IpaH7.8 targets human (but not mouse) GSDMD through a similar mechanism. The structure of full-length GSDMB suggests stronger autoinhibition than in other gasdermins9,10. GSDMB has multiple splicing isoforms that are equally targeted by IpaH7.8 but exhibit contrasting pyroptotic activities. Presence of exon 6 in the isoforms dictates the pore-forming, pyroptotic activity in GSDMB. We determine the cryo-electron microscopy structure of the 27-fold-symmetric GSDMB pore and depict conformational changes that drive pore formation. The structure uncovers an essential role for exon-6-derived elements in pore assembly, explaining pyroptosis deficiency in the non-canonical splicing isoform used in recent studies6,8. Different cancer cell lines have markedly different isoform compositions, correlating with the onset and extent of pyroptosis following GZMA stimulation. Our study illustrates fine regulation of GSDMB pore-forming activity by pathogenic bacteria and mRNA splicing and defines the underlying structural mechanisms.


Asunto(s)
Gasderminas , Proteínas Citotóxicas Formadoras de Poros , Animales , Humanos , Ratones , Línea Celular Tumoral , Microscopía por Crioelectrón , Cristalografía por Rayos X , Gasderminas/química , Gasderminas/genética , Gasderminas/metabolismo , Gasderminas/ultraestructura , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/ultraestructura , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestructura , Piroptosis , Shigella flexneri , Especificidad de la Especie , Empalme Alternativo
10.
J Asthma ; 60(9): 1751-1760, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36847643

RESUMEN

BACKGROUND: Allergic rhinitis (AR) is a great risk factor for developing asthma, and its pathogenesis is affected by various factors, such as gene and environment. GSDMB is related to allergic diseases. Our purpose is to explore the correlation of single nucleotide polymorphisms (SNPs) in GSDMB and AR risk in the Chinese population. METHODS: We performed a case-control study including 1005 cases and 1004 controls. Rs2305479, rs4795400, and rs12450091 in GSDMB were geneotyped using Agena MassARRAY. The relationships between GSDMB SNPs and AR risk were assessed by logistic regression analysis in PLINK1.9. RESULTS: Our study showed that rs4795400 was a protective factor for AR in overall (TT vs. CC: OR = 0.66, p = 0.009; TT vs. CC/TC: OR = 0.67, p = 0.008; additive: OR = 0.87, p = 0.042 males, people with BMI ≤ 24, and living in wind-blown sand area. Rs2305479 was associated with a reduced AR risk in males (TT vs. CC: OR = 0.47, p = 0.014; TT vs. CC/TC: OR = 0.43, p = 0.004). However, rs12450091 was a risk factor for AR in people living in the loess hilly region (CC: OR = 4.75, p = 0.047). The levels of EO and EO_per in the case group were significantly higher than those in the control group (p < 0.05). CONCLUSION: This study indicated that GSDMB polymorphisms (rs4795400, rs2305479, and rs12450091) were associated with AR susceptibility. Further studies are required to confirm our findings and to clarify the functional relationship.


Asunto(s)
Asma , Rinitis Alérgica , Masculino , Humanos , Genotipo , Predisposición Genética a la Enfermedad , Estudios de Casos y Controles , Pueblos del Este de Asia , Asma/epidemiología , Asma/genética , Rinitis Alérgica/epidemiología , Rinitis Alérgica/genética , Polimorfismo de Nucleótido Simple , Proteínas Citotóxicas Formadoras de Poros/genética
11.
Nat Commun ; 14(1): 852, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36792583

RESUMEN

The vitamin D binding protein (DBP), encoded by the group-specific component (GC) gene, is a component of the vitamin D system. In a genome-wide association study of DBP concentration in 65,589 neonates we identify 26 independent loci, 17 of which are in or close to the GC gene, with fine-mapping identifying 2 missense variants on chromosomes 12 and 17 (within SH2B3 and GSDMA, respectively). When adjusted for GC haplotypes, we find 15 independent loci distributed over 10 chromosomes. Mendelian randomization analyses identify a unidirectional effect of higher DBP concentration and (a) higher 25-hydroxyvitamin D concentration, and (b) a reduced risk of multiple sclerosis and rheumatoid arthritis. A phenome-wide association study confirms that higher DBP concentration is associated with a reduced risk of vitamin D deficiency. Our findings provide valuable insights into the influence of DBP on vitamin D status and a range of health outcomes.


Asunto(s)
Estudio de Asociación del Genoma Completo , Proteína de Unión a Vitamina D , Recién Nacido , Humanos , Proteína de Unión a Vitamina D/genética , Vitamina D/genética , Calcifediol , Vitaminas , Polimorfismo de Nucleótido Simple , Proteínas Citotóxicas Formadoras de Poros/genética
12.
Front Immunol ; 13: 893914, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36217543

RESUMEN

Background: Gasdermin D (GSDMD) plays an essential role in the pathway of pyroptosis. However, whether GSDMD participates in myocardial ischaemia/reperfusion injury (MI/RI) remains poorly understood. Methods: Serum levels of GSDMD and IL-18 in ST-segment elevation myocardial infarction (STEMI) patients were measured by ELISA. The expression of GSDMD and GSDMD N-terminal (GSDMD-NT) in vivo and in vitro was assessed by western blot and immunofluorescence staining. GSDMD-/- mice and wild type (WT) mice were induced MI/RI, followed by cardiac ultrasound and histological analysis. Results: Clinically, patients suffering from STEMI after percutaneous coronary intervention (PCI) exhibited higher levels of GSDMD and IL-18 than that in the controls. In vitro, the cleavage of GSDMD was significantly upregulated in macrophages exposed to hypoxia/reoxygenation or H2O2. In vivo, the levels of GSDMD and GSDMD-NT increased notably after MI/RI, especially in macrophages infiltrating in the infarct area. Moreover, compared with WT mice, GSDMD-/- mice showed reduced infarct size (25.45 ± 3.07% versus 36.47 ± 3.72%), improved left ventricular ejection fraction (37.71 ± 1.81% versus 29.44 ± 2.28%) and left ventricular fractional shortening (18.01 ± 0.97% versus 13.62 ± 1.15%) as well as attenuated pathological damage after I/R injury, along with reduced levels of proinflammatory cytokines and decreased infiltration of neutrophils. Conclusions: Our study revealed that GSDMD deficiency significantly alleviated the inflammatory response by regulating pyroptosis, reduced the infarct size and preserved cardiac function after MI/RI, thus providing a potential strategy for the treatment of myocardial reperfusion injury.


Asunto(s)
Daño por Reperfusión Miocárdica , Intervención Coronaria Percutánea , Proteínas de Unión a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Infarto del Miocardio con Elevación del ST , Animales , Citocinas/metabolismo , Peróxido de Hidrógeno/metabolismo , Interleucina-18/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Piroptosis , Volumen Sistólico , Función Ventricular Izquierda
13.
PLoS One ; 17(10): e0274951, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36201519

RESUMEN

BACKGROUND: Gasdermin A (GSDMA) and gasdermin B (GSDMB) have been associated with childhood and adult asthma in many populations including the Jordanian population. It is also known that IgE plays a crucial role in various allergic disorders, such elevated levels of total serum IgE were detected in asthma and allergic rhinitis. IgE immunoglobulin is responsible for the release of numerous inflammatory mediators, such as histamine and prostaglandins, from mast cells in asthmatic patients. OBJECTIVE: In this study, single nucleotide polymorphisms of GSDMA (rs7212938, T/G) and GSDMB (rs7216389, T/C) in Jordanian population were investigated for their association with total IgE levels in serum of asthmatic children and adult subjects. METHODS: The genetic polymorphism analysis for SNPs was performed using the polymerase chain reaction (PCR)/restriction fragment length polymorphism method (RFLP). Three analysis models were applied to the genotype data: co-dominant, dominant and recessive. RESULTS: Our data demonstrate a significant correlation between GSDMB genetic SNP (rs7216389) and the total IgE serum level. Where one minor allele in the GSDMB gene is sufficient to induce significant changes in the IgE serum levels and plays a role in the pathogenesis of asthma in asthmatic children of the Jordanian population. Suggesting that this polymorphism might have a protective effect against asthma risk. While the presence of the GSDMB polymorphism alone might not be sufficient to associate with the high risk of developing asthma or responding to it in adults in Jordanian population. CONCLUSION: In conclusion, the current study confirms the significant association of GSDMB genetic SNP (rs7216389) with IgE levels in asthma patients in Jordanian population, while no significant correlation of GSDMA and IgE level was found in both child and adult asthmatic patients.


Asunto(s)
Asma , Predisposición Genética a la Enfermedad , Proteínas Citotóxicas Formadoras de Poros/genética , Adulto , Asma/genética , Estudios de Casos y Controles , Niño , Genotipo , Histamina , Humanos , Inmunoglobulina E , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Prostaglandinas
14.
Mediators Inflamm ; 2022: 7853482, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36065376

RESUMEN

The adipose tissue NLRP3 inflammasome has recently emerged as a contributor to obesity-related metabolic inflammation. Recent studies have demonstrated that the activation of the NLRP3 inflammasome cleaves gasdermin D (GSDMD) and induces pyroptosis, a proinflammatory programmed cell death. However, whether GSDMD is involved in the regulation of adipose tissue function and the development of obesity-induced metabolic disease remains unknown. The aim of the present study was to investigate the role of GSDMD in adipose tissue inflammation as well as whole-body metabolism using GSDMD-deficient mice fed a high-fat diet (HFD) for 30 weeks. The effects of GSDMD deficiency on adipose tissue, liver, and isolated macrophages from wild-type (WT) and GSDMD knockout (KO) mice were examined. In addition, 3T3-L1 cells were used to examine the expression of GSDMD during adipogenesis. The results demonstrate that although HFD-induced inflammation was partly ameliorated in isolated macrophages and liver, adipose tissue remained unaffected by GSDMD deficiency. Compared with the WT HFD mice, GSDMD KO HFD mice exhibited a mild increase in HFD-induced glucose intolerance with increased systemic and adipose tissue IL-1ß levels. Interestingly, GSDMD deficiency caused accumulation of fat mass when challenged with HFD, partly by suppressing the expression of peroxisome proliferator-activated receptor gamma (PPARγ). The expression of GSDMD mRNA and protein was dramatically suppressed during adipocyte differentiation and was inversely correlated with PPARγ expression. Together, these findings indicate that GSDMD is not a prerequisite for HFD-induced adipose tissue inflammation and suggest a noncanonical function of GSDMD in regulation of fat mass through PPARγ.


Asunto(s)
Tejido Adiposo , Dieta Alta en Grasa , Intolerancia a la Glucosa , Proteínas de Unión a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Obesidad/metabolismo , PPAR gamma/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas Citotóxicas Formadoras de Poros/genética
15.
J Biol Chem ; 298(10): 102441, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36055404

RESUMEN

Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging ß-barrel pore-forming toxin. Upon binding to the target membranes, VCC monomers first assemble into oligomeric prepore intermediates and subsequently transform into transmembrane ß-barrel pores. VCC harbors a designated pore-forming motif, which, during oligomeric pore formation, inserts into the membrane and generates a transmembrane ß-barrel scaffold. It remains an enigma how the molecular architecture of the pore-forming motif regulates the VCC pore-formation mechanism. Here, we show that a specific pore-forming motif residue, E289, plays crucial regulatory roles in the pore-formation mechanism of VCC. We find that the mutation of E289A drastically compromises pore-forming activity, without affecting the structural integrity and membrane-binding potential of the toxin monomers. Although our single-particle cryo-EM analysis reveals WT-like oligomeric ß-barrel pore formation by E289A-VCC in the membrane, we demonstrate that the mutant shows severely delayed kinetics in terms of pore-forming ability that can be rescued with elevated temperature conditions. We find that the pore-formation efficacy of E289A-VCC appears to be more profoundly dependent on temperature than that of the WT toxin. Our results suggest that the E289A mutation traps membrane-bound toxin molecules in the prepore-like intermediate state that is hindered from converting into the functional ß-barrel pores by a large energy barrier, thus highlighting the importance of this residue for the pore-formation mechanism of VCC.


Asunto(s)
Proteínas Bacterianas , Citotoxinas , Proteínas Citotóxicas Formadoras de Poros , Vibrio cholerae , Factores de Virulencia , Membrana Celular/metabolismo , Citotoxinas/química , Citotoxinas/genética , Vibrio cholerae/patogenicidad , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Factores de Virulencia/química , Factores de Virulencia/genética , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Secuencias de Aminoácidos , Mutación , Ácido Glutámico/química , Ácido Glutámico/genética
16.
Autoimmunity ; 55(8): 587-596, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35993279

RESUMEN

Acute lung injury (ALI) is considered as a severe respiratory disease with aggravated inflammatory responses. Krüppel-like factor 9 (KLF9), a member of KLF family, has been reported to be involved in inflammatory disorders. However, the effect of KLF9 in ALI has not been elucidated. Here the present study was to clarify the role of KLF9 and its mechanism in ALI. The ALI in vitro model was established with lipopolysaccharide (LPS)-treated RAW264.7 cells. Mice were injected with LPS to conduct an ALI in vivo model. The expression of KLF9 and gasdermin D (GSDMD) was examined using quantitative reverse transcription-PCR, haematoxylin-eosin/immunohistochemistry staining and western blot assays. Enzyme-linked immunosorbent assay was employed to detect the levels of inflammatory cytokines. JASPAR database was used to predict the recognition motif of KLF9, and the relationship between KLF9 and GSDMD was determined by luciferase reporter assay and chromatin immunoprecipitation analysis. The number of neutrophils in bronchoalveolar lavage fluid as well as the wet/dry weight ratio was caculated. The results showed that The expression of KLF9 in lung was significantly increased in LPS-stimulated mice. Moreover, KLF9 knockout relieved the lung injury in ALI mice. GSDMD is one of targets genes of the transcription factor KLF9. KLF9 knockout induced a decreased expression of GSDMD in LPS-treated mice. Furthermore, in RAW264.7 cells after LPS administration, KLF9 knockdown reduced the levels of inflammatory factors and suppressed the expression of GSDMD. In summarise, these findings exhibited that KLF9 knockout could mitigate the lung injury and inflammatory responses in ALI mice by directly regulating GSDMD.


Asunto(s)
Lesión Pulmonar Aguda , Factores de Transcripción de Tipo Kruppel , Proteínas de Unión a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Animales , Citocinas/metabolismo , Regulación hacia Abajo , Inflamación/genética , Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Ratones , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Células RAW 264.7
17.
Clin Transl Med ; 12(8): e1002, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36030524

RESUMEN

BACKGROUNDS: Inflammation underlies the mechanism of different kinds of heart disease. Cytoplasmic membrane localized N-terminal fragment of gasdermin-D (GSDMD-N) could induce inflammatory injury to cardiomyocyte. However, effects and dynamic changes of GSDMD during the process of lipopolysaccharide (LPS) related inflammatory stress induced cardiomyocyte injury are barely elucidated to date. In this study, LPS related cardiomyocyte injury was investigated based on potential interaction of GSDMD-N induced mitochondrial injury and mitophagy mediated mitochondria quality control. METHODS: HL-1 cardiomyocytes were treated with LPS and Nigericin to induce inflammatory stress. The dual-fluorescence-labelled GSDMD expressed HL-1 cardiomyocytes were constructed to study the translocation of GSDMD. The mitochondrial membrane potential (MMP) was measured by JC-1 staining. Mitophagy and autophagic flux were recorded by transmission electron microscopy and fluorescent image. RESULTS: GSDMD-N showed a time-dependent pattern of translocation from mitochondria to cytoplasmic membrane under LPS and Nigericin induced inflammatory stress in HL-1 cardiomyocytes. GSDMD-N preferred to localize to mitochondria to permeablize its membrane and dissipate the MMP. This effect couldn't be reversed by cyclosporine-A (mPTP inhibitor), indicating GSDMD-N pores as alternative mechanism underlying MMP regulation, in addition to mitochondrial permeability transition pore (mPTP). Moreover, the combination between GSDMD-N and autophagy related Microtubule Associated Protein 1 Light Chain 3 Beta (LC3B) was verified by co-immunoprecipitation. Besides, mitophagy alleviating GSDMD-N induced mitochondrial injury was proved by pre-treatment of autophagy antagonist or agonist in GSDMD-knock out or GSDMD-overexpression cells. A time-dependent pattern of GSDMD translocation and mitochondrial GSDMD targeted mitophagy were verified. CONCLUSION: Herein, our study confirmed a crosstalk between GSDMD-N induced mitochondrial injury and mitophagy mediated mitochondria quality control during LPS related inflammation induced cardiomyocyte injury, which potentially facilitating the development of therapeutic target to myocardial inflammatory disease. Our findings support pharmaceutical intervention on enhancing autophagy or inhibiting GSDMD as potential target for inflammatory heart disease treatment.


Asunto(s)
Cardiopatías , Mitocondrias , Mitofagia , Miocitos Cardíacos , Proteínas de Unión a Fosfato , Proteínas Citotóxicas Formadoras de Poros , Humanos , Inflamación , Lipopolisacáridos , Mitocondrias/patología , Poro de Transición de la Permeabilidad Mitocondrial , Miocitos Cardíacos/efectos de los fármacos , Nigericina , Proteínas de Unión a Fosfato/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Control de Calidad
18.
Proc Natl Acad Sci U S A ; 119(31): e2123017119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35881802

RESUMEN

Staphylococcus aureus is an opportunistic pathogen and chief among bloodstream-infecting bacteria. S. aureus produces an array of human-specific virulence factors that may contribute to immune suppression. Here, we defined the response of primary human phagocytes following infection with S. aureus using RNA-sequencing (RNA-Seq). We found that the overall transcriptional response to S. aureus was weak both in the number of genes and in the magnitude of response. Using an ex vivo bacteremia model with fresh human blood, we uncovered that infection with S. aureus resulted in the down-regulation of genes related to innate immune response and cytokine and chemokine signaling. This muted transcriptional response was conserved across diverse S. aureus clones but absent in blood exposed to heat-killed S. aureus or blood infected with the less virulent staphylococcal species Staphylococcus epidermidis. Notably, this signature was also present in patients with S. aureus bacteremia. We identified the master regulator S. aureus exoprotein expression (SaeRS) and the SaeRS-regulated pore-forming toxins as key mediators of the transcriptional suppression. The S. aureus-mediated suppression of chemokine and cytokine transcription was reflected by circulating protein levels in the plasma. Wild-type S. aureus elicited a soluble milieu that was restrictive in the recruitment of human neutrophils compared with strains lacking saeRS. Thus, S. aureus blunts the inflammatory response resulting in impaired neutrophil recruitment, which could promote the survival of the pathogen during invasive infection.


Asunto(s)
Interacciones Huésped-Patógeno , Neutrófilos , Infecciones Estafilocócicas , Staphylococcus aureus , Bacteriemia/inmunología , Bacteriemia/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citocinas/metabolismo , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Neutrófilos/inmunología , Neutrófilos/microbiología , Proteínas Citotóxicas Formadoras de Poros/genética , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Staphylococcus epidermidis/patogenicidad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Am J Respir Cell Mol Biol ; 67(4): 430-437, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35580164

RESUMEN

Chromosome 17q12-q21 is the most replicated genetic locus for childhood-onset asthma. Polymorphisms in this locus containing ∼10 genes interact with a variety of environmental exposures in the home and outdoors to modify asthma risk. However, the functional basis for these associations and their linkages to the environment have remained enigmatic. Within this extended region, regulation of GSDMB (gasdermin B) expression in airway epithelial cells has emerged as the primary mechanism underlying the 17q12-q21 genome-wide association study signal. Asthma-associated SNPs influence the abundance of GSDMB transcripts as well as the functional properties of GSDMB protein in airway epithelial cells. GSDMB is a member of the gasdermin family of proteins, which regulate pyroptosis and inflammatory responses to microbial infections. The aims of this review are to synthesize recent studies on the relationship of 17q12-q21 SNPs to childhood asthma and the evidence pointing to GSDMB gene expression or protein function as the underlying mechanism and to explore the potential functions of GSDMB that may influence the risk of developing asthma during childhood.


Asunto(s)
Asma , Estudio de Asociación del Genoma Completo , Proteínas Citotóxicas Formadoras de Poros/genética , Asma/genética , Asma/metabolismo , Sitios Genéticos , Predisposición Genética a la Enfermedad , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polimorfismo de Nucleótido Simple
20.
Acta Otolaryngol ; 142(5): 448-453, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35640035

RESUMEN

BACKGROUND: To date, seven DFNA5 mutations have been reported in families with autosomal dominant non-syndromic hearing loss worldwide. All the mutations cause exon 8 skipping at the mRNA level, that led to the protein truncated and the protein could exert a gain of ototoxic function. OBJECTIVE: In this study, we found an autosomal-dominant non-syndromic hearing loss Chinese pedigree which spanned four generations and comprised 43 members. We want to identify the causative gene and mutation. METHODS: Application of microsatellite markers on DFNA 23 loci preliminary screening of 25 genes, data were analyzed by linkage analysis. RESULTS: We mapped the locus to the region between D7S629 and D7S516 (two-point lod-score of 5.39) with the application of 8 microsatellite markers. By direct sequencing of candidate genes in mapping region, we identified a novel missense mutation ivs7-2 A > G in DFNA5 gene, which was faithfully cosegregated with hearing loss in the family. CONCLUSION AND SIGNIFICANCE: The missense mutation in intron 7 of DFNA5 causes skipping of exon 8, resulting in premature termination of the open reading frame. This type of mutation has repeatedly confirmed that it provides more evidence for the previous view and provides a more solid foundation for future research.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Proteínas Citotóxicas Formadoras de Poros , Humanos , China , Sordera/genética , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Mutación , Linaje , Proteínas Citotóxicas Formadoras de Poros/genética , Receptores de Estrógenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA