Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 276
Filter
1.
J Biol Chem ; 300(4): 107200, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38508315

ABSTRACT

Interferon (IFN) regulatory factors (IRF) are key transcription factors in cellular antiviral responses. IRF7, a virus-inducible IRF, expressed primarily in myeloid cells, is required for transcriptional induction of interferon α and antiviral genes. IRF7 is activated by virus-induced phosphorylation in the cytoplasm, leading to its translocation to the nucleus for transcriptional activity. Here, we revealed a nontranscriptional activity of IRF7 contributing to its antiviral functions. IRF7 interacted with the pro-inflammatory transcription factor NF-κB-p65 and inhibited the induction of inflammatory target genes. Using knockdown, knockout, and overexpression strategies, we demonstrated that IRF7 inhibited NF-κB-dependent inflammatory target genes, induced by virus infection or toll-like receptor stimulation. A mutant IRF7, defective in transcriptional activity, interacted with NF-κB-p65 and suppressed NF-κB-induced gene expression. A single-action IRF7 mutant, active in anti-inflammatory function, but defective in transcriptional activity, efficiently suppressed Sendai virus and murine hepatitis virus replication. We, therefore, uncovered an anti-inflammatory function for IRF7, independent of transcriptional activity, contributing to the antiviral response of IRF7.


Subject(s)
Interferon Regulatory Factor-7 , NF-kappa B , Animals , Humans , Mice , HEK293 Cells , Inflammation/genetics , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Sendai virus/physiology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Virus Replication , Mutation , Gene Expression Regulation/genetics , Murine hepatitis virus/physiology , Coronavirus Infections/immunology , Respirovirus Infections/immunology
3.
Int Immunopharmacol ; 102: 108382, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34848155

ABSTRACT

Nephrotoxicity is an indication for the damage of kidney-specific detoxification and excretion mechanisms by exogenous or endogenous toxicants. Exposure to vancomycin predominantly results in renal damage and losing the control of body homeostasis. Vancomycin-treated rats (200 mg/kg/once daily, for seven consecutive days, i.p.) revealed significant increase in serum pivotal kidney function, oxidative stress, and inflammatory biomarkers. Histologically, vancomycin showed diffuse acute tubular necrosis, denudation of epithelium and infiltration of inflammatory cells in the lining tubular epithelium in cortical portion. In the existing study, the conservative consequences of scopoletin against vancomycin nephrotoxicity was investigated centering on its capacity to alleviate oxidative strain and inflammation through streamlining nuclear factor (erythroid-derived-2) like 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling and prohibiting the nuclear factor kappa B (NF-κB)/mitogen-activated protein kinase (p38 MAPK) pathway. With respect to vancomycin group, scopoletin pretreatment (50 mg/kg/once daily, i.p.) efficiently reduced kidney function, oxidative stress biomarkers and inflammatory mediators. Moreover, histological and immunohistochemical examination of scopoletin-treated group showed remarkable improvement in histological structure and reduced vancomycin-induced renal expression of iNOS, NF-κB and p38 MAPK. In addition, scopoletin downregulated (Kelch Like ECH Associated Protein1) Keap1, P38MAPK and NF-κB expression levels while upregulated renal expression levels of regulatory protein (IκBα), Nrf2 and HO-1. Furthermore, molecular docking and network approach were constructed to study the prospect interaction between scopoletin and the targeted proteins that streamline oxidative stress and inflammatory pathways. The present investigations elucidated that scopoletin co-treatment with vancomycin may be a rational curative protocol for mitigation of vancomycin-induced renal intoxication.


Subject(s)
Anti-Bacterial Agents , Kidney Diseases/drug therapy , Protective Agents/therapeutic use , Scopoletin/therapeutic use , Signal Transduction/drug effects , Vancomycin , Animals , Cytokines/blood , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/immunology , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/immunology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/immunology , Kidney Diseases/pathology , Male , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/immunology , Nitric Oxide Synthase Type II/immunology , Protective Agents/pharmacology , Rats, Wistar , Scopoletin/pharmacology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
4.
Sci Rep ; 11(1): 22913, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824310

ABSTRACT

Inflammation is a physiological process whose deregulation causes some diseases including cancer. Nuclear Factor kB (NF-kB) is a family of ubiquitous and inducible transcription factors, in which the p65/p50 heterodimer is the most abundant complex, that play critical roles mainly in inflammation. Glucocorticoid Receptor (GR) is a ligand-activated transcription factor and acts as an anti-inflammatory agent and immunosuppressant. Thus, NF-kB and GR are physiological antagonists in the inflammation process. Here we show that in mice and humans there is a spliced variant of p65, named p65 iso5, which binds the corticosteroid hormone dexamethasone amplifying the effect of the glucocorticoid receptor and is expressed in the liver of patients with hepatic cirrhosis and hepatocellular carcinoma (HCC). Furthermore, we have quantified the gene expression level of p65 and p65 iso5 in the PBMC of patients affected by SARS-CoV-2 disease. The results showed that in these patients the p65 and p65 iso5 mRNA levels are higher than in healthy subjects. The ability of p65 iso5 to bind dexamethasone and the regulation of the glucocorticoid (GC) response in the opposite way of the wild type improves our knowledge and understanding of the anti-inflammatory response and identifies it as a new therapeutic target to control inflammation and related diseases.


Subject(s)
Inflammation/immunology , Receptors, Glucocorticoid/metabolism , Transcription Factor RelA/metabolism , Adrenal Cortex Hormones/metabolism , Adult , Alternative Splicing , Animals , COVID-19/immunology , Carcinoma, Hepatocellular/metabolism , Dexamethasone/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Glucocorticoids/metabolism , Hepatitis/metabolism , Humans , Inflammation/metabolism , Leukocytes, Mononuclear/metabolism , Liver/metabolism , Liver Diseases/immunology , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , NF-kappa B/metabolism , Protein Isoforms , Receptors, Glucocorticoid/immunology , SARS-CoV-2/pathogenicity , Transcription Factor RelA/immunology , Transcription Factor RelA/physiology
5.
Front Immunol ; 12: 749646, 2021.
Article in English | MEDLINE | ID: mdl-34712238

ABSTRACT

The nucleotide-binding leucine-rich repeat-containing receptor (NLR) family protein-5 (NLRC5) controls NF-κB activation and production of inflammatory cytokines in certain cell types. NLRC5 is considered a potential regulator of hepatic fibrogenic response due to its ability to inhibit hepatic stellate activation in vitro. To test whether NLRC5 is critical to control liver fibrosis, we treated wildtype and NLRC5-deficient mice with carbon tetrachloride (CCl4) and assessed pathological changes in the liver. Serum alanine transaminase levels and histopathology examination of liver sections revealed that NLRC5 deficiency did not exacerbate CCl4-induced liver damage or inflammatory cell infiltration. Sirius red staining of collagen fibers and hydroxyproline content showed comparable levels of liver fibrosis in CCl4-treated NLRC5-deficient and control mice. Myofibroblast differentiation and induction of collagen genes were similarly increased in both groups. Strikingly, the fibrotic livers of NLRC5-deficient mice showed reduced expression of matrix metalloproteinase-3 (Mmp3) and tissue inhibitor of MMPs-1 (Timp1) but not Mmp2 or Timp2. Fibrotic livers of NLRC5-deficient mice had increased expression of TNF but similar induction of TGFß compared to wildtype mice. CCl4-treated control and NLRC5-deficient mice displayed similar upregulation of Cx3cr1, a monocyte chemoattractant receptor gene, and the Cd68 macrophage marker. However, the fibrotic livers of NLRC5-deficient mice showed increased expression of F4/80 (Adgre1), a marker of tissue-resident macrophages. NLRC5-deficient livers showed increased phosphorylation of the NF-κB subunit p65 that remained elevated following fibrosis induction. Taken together, NLRC5 deficiency deregulates hepatic inflammatory response following chemical injury but does not significantly aggravate the fibrogenic response, showing that NLRC5 is not a critical regulator of liver fibrosis pathogenesis.


Subject(s)
Intracellular Signaling Peptides and Proteins/immunology , Liver Cirrhosis/immunology , Alanine Transaminase/blood , Animals , Carbon Tetrachloride , Cytokines/blood , Cytokines/genetics , Cytokines/immunology , Gene Expression , Intracellular Signaling Peptides and Proteins/genetics , Liver/immunology , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Transcription Factor RelA/immunology
6.
Int Immunopharmacol ; 99: 107918, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34320458

ABSTRACT

OBJECT: Ninjurin2 (nerve injury induced protein 2, NINJ2) is a molecule which mediates cell-to-cell and cell-to-extracellular matrix interactions in the nervous system. Clinical study shows NINJ2 is associated with the development of postherpetic neuralgia. However, it is lack of direct evidence that NINJ2 participated in neuropathic pain. In this study, we aim to investigate the role of NINJ2 in the development of neuropathic pain in spared sciatic nerve injury rats and the underlying mechanism. METHOD: Spared sciatic nerve injury (SNI) models were established. The level of NINJ2 and p-p65 (a NF-κB family member) were measured in SNI rats by western blots and immunofluorescent staining. Lentivirus encoding small interfering RNA targeting NINJ2 (RNAi) was intrathecally injected into rats. Then the change of pain behavior of rats induced by NINJ2 RNAi was tested by Von-Frey hairs. The change of p-p65 in the spinal cord in rats after NINJ2 RNAi treatment was also measured by western blots. inhibitor of p-p65-induced change of TNF-α, IL-1ß, and IL-6 levels were measured by ELISA. RESULTS: NINJ2 and p-p65 were increased in the spinal cord of SNI rats on the 3, 7, 14th days after modeling. NINJ2 were mainly expressed in neurons, and co-located with p-p65 in the spinal dorsal horn. When down regulating the level of NINJ2 by RNAi, the development of pain in SNI rats was partially blocked. Phosphorylation of p65 was also inhibited by NINJ2 RNAi. Blocking the phosphorylation of NF-κB pathway could inhibit the increase of TNF-α, IL-1ß, and IL-6 in the spinal cord of SNI rats. CONCLUSION: NINJ2 protein was increased in the spinal cord of SNI rats. It participated in the development of nerve injury-induced neuropathic pain by activating neuroinflammation in the spinal cord via NF-κB pathway. This study provides a new target to investigate the mechanism of neuropathic pain.


Subject(s)
Cell Adhesion Molecules, Neuronal/immunology , Neuralgia/immunology , Neuroinflammatory Diseases/immunology , Sciatic Nerve/injuries , Transcription Factor RelA/immunology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Male , Rats, Sprague-Dawley , Sciatic Nerve/immunology , Spinal Cord/immunology
7.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070750

ABSTRACT

The immune system is a fine modulator of the tumor biology supporting or inhibiting its progression, growth, invasion and conveys the pharmacological treatment effect. Tumors, on their side, have developed escaping mechanisms from the immune system action ranging from the direct secretion of biochemical signals to an indirect reaction, in which the cellular actors of the tumor microenvironment (TME) collaborate to mechanically condition the extracellular matrix (ECM) making it inhospitable to immune cells. TME is composed of several cell lines besides cancer cells, including tumor-associated macrophages, cancer-associated fibroblasts, CD4+ and CD8+ lymphocytes, and innate immunity cells. These populations interface with each other to prepare a conservative response, capable of evading the defense mechanisms implemented by the host's immune system. The presence or absence, in particular, of cytotoxic CD8+ cells in the vicinity of the main tumor mass, is able to predict, respectively, the success or failure of drug therapy. Among various mechanisms of immunescaping, in this study, we characterized the modulation of the phenotypic profile of CD4+ and CD8+ cells in resting and activated states, in response to the mechanical pressure exerted by a three-dimensional in vitro system, able to recapitulate the rheological and stiffness properties of the tumor ECM.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Extracellular Matrix/immunology , Gene Expression Regulation, Neoplastic/immunology , Tumor Escape , Tumor Microenvironment/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cell Culture Techniques , Elastic Modulus , Extracellular Matrix/chemistry , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Humans , Hydrogels/chemistry , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Mechanotransduction, Cellular , Models, Biological , NF-kappa B/genetics , NF-kappa B/immunology , Phenotype , Primary Cell Culture , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Rheology , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
8.
Front Immunol ; 12: 644862, 2021.
Article in English | MEDLINE | ID: mdl-34093533

ABSTRACT

NLRP3 inflammasome has emerged as a crucial regulator of inflammatory bowel disease (IBD) characterized by a chronic inflammatory disease of the gastrointestinal tract. The expression of MCT4 is significantly increased in intestinal mucosal tissue of IBD, which has been identified to regulate intestinal barrier function. However, the function of MCT4 in cell pyroptosis remained unknown. In this study, we have established a stable cell line with MCT4 overexpression in HT-29 and CaCO2 cells, respectively. Functional analysis revealed that ectopic expression of MCT4 in CaCO2 cells contributed to cell pyroptosis as evidenced by LDH assay, which is largely attributed to Caspase-1-mediated canonical pyroptosis, but not Caspase-4 and Caspase-5, leading to cleave pro-IL-1ß and IL-18 into mature form and release mediated by cleaved GSDMD. Mechanically, MCT4 overexpression in HT-29 and CaCO2 cell triggered the phosphorylation of ERK1/2 and NF-κB p65, while inhibition of MCT4 by MCT inhibitor α-Cyano-4-hydroxycinnamic acid (α-CHCA) in HT-29 and CaCO2 cells led to a significant downregulation of ERK1/2 and NF-κB activity. What's more, blockade of ERK1/2-NF-κB pathway could reverse the promotion effect of MCT4 on IL-1ß expression. Importantly, both MCT4 and Caspase-1, GSDMD were significantly increased in patients with IBD, and a positive clinical correlation between MCT4 and Caspase-1 expression was observed (p < 0.001). Taken together, these findings suggested that MCT4 promoted Caspase-1-mediated canonical cell pyroptosis to aggravate intestinal inflammation in intestinal epithelial cells (IECs) through the ERK1/2-NF-κB pathway.


Subject(s)
Inflammatory Bowel Diseases/immunology , MAP Kinase Signaling System/immunology , Monocarboxylic Acid Transporters/immunology , Muscle Proteins/immunology , Pyroptosis/immunology , Caco-2 Cells , Caspases/immunology , HT29 Cells , Humans , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Interleukin-18/immunology , Interleukin-1beta/immunology , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , Transcription Factor RelA/immunology
9.
Clin Exp Immunol ; 206(1): 110-117, 2021 10.
Article in English | MEDLINE | ID: mdl-34118070

ABSTRACT

Guillain-Barré syndrome (GBS) is an acute inflammatory and immune-mediated demyelinating disease of the peripheral nervous system (PNS). Macrophages play a central role in its animal model, experimental autoimmune neuritis (EAN), which has been well accepted. Additionally, nuclear factor (NF)-κB inhibitors have been used to treat cancers and have shown beneficial effects. Here, we investigated the therapeutic effect of M2 macrophage and the NF-κB pathway's correlation with macrophage activation in EAN in C57BL/6 mice. We demonstrate that M2 macrophage transfusion could alleviate the clinical symptoms of EAN by reducing the proportion of M1 macrophage in the peak period, inhibiting the phosphorylation of NF-κB p65. The NF-κB inhibitor (BAY-11-7082) could alleviate the clinical symptoms of EAN and shorten the duration of symptoms by reducing the proportion of M1 macrophages and the expression of proinflammatory cytokines. Consequently, BAY-11-7082 exhibits strong potential as a therapeutic strategy for ameliorating EAN by influencing the balance of M1/M2 macrophages and inflammatory cytokines.


Subject(s)
Guillain-Barre Syndrome , Macrophages/immunology , Neuritis, Autoimmune, Experimental , Nitriles/pharmacology , Sulfones/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Animals , Guillain-Barre Syndrome/drug therapy , Guillain-Barre Syndrome/immunology , Male , Mice , Neuritis, Autoimmune, Experimental/drug therapy , Neuritis, Autoimmune, Experimental/immunology , Transcription Factor RelA/immunology
10.
J Immunol ; 206(12): 2852-2861, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34049972

ABSTRACT

NF-κB plays a crucial role in regulating cell proliferation, inflammation, apoptosis, and immune responses. HSV type 2 (HSV-2) is one of the most predominant sexually transmitted pathogens worldwide, and its infection increases the risk of HIV type 1 (HIV-1) acquisition and transmission. HSV-2 glycoprotein D (gD), highly homologous to HSV-1 gD, is essential for viral adhesion, fusion, entry, and spread. It is known that HSV-1 gD can bind herpesvirus entry mediator (HVEM) to trigger NF-κB activation and thereby facilitate viral replication at the early stage of infection. In this study, we found that purified HSV-2 gD triggered NF-κB activation at the early stage of infection, whereas ectopic expression of HSV-2 gD significantly downregulated TNF-α-induced NF-κB activity as well as TNF-α-induced IL-6 and IL-8 expression. Mechanistically, HSV-2 gD inhibited NF-κB, but not IFN-regulatory factor 3 (IRF3), activation and suppressed NF-κB activation mediated by overexpression of TNFR-associated factor 2 (TRAF2), IκB kinase α (IKKα), IKKß, or p65. Coimmunoprecipitation and binding kinetic analyses demonstrated that HSV-2 gD directly bound to the NF-κB subunit p65 and abolished the nuclear translocation of p65 upon TNF-α stimulation. Mutational analyses further revealed that HSV-2 gD interacted with the region spanning aa 19-187 of p65. Findings in this study together demonstrate that HSV-2 gD interacts with p65 to regulate p65 subcellular localization and thereby prevents NF-κB-dependent gene expression, which may contribute to HSV-2 immune evasion and pathogenesis.


Subject(s)
Herpesvirus 2, Human/immunology , Transcription Factor RelA/immunology , Viral Envelope Proteins/immunology , HEK293 Cells , HeLa Cells , Humans
11.
Viruses ; 13(2)2021 01 31.
Article in English | MEDLINE | ID: mdl-33572560

ABSTRACT

The innate immune response (IIR) involves rapid genomic expression of protective interferons (IFNs) and inflammatory cytokines triggered by intracellular viral replication. Although the transcriptional control of the innate pathway is known in substantial detail, little is understood about the complexity of alternative splicing (AS) and alternative polyadenylation (APA) of mRNAs underlying the cellular IIR. In this study, we applied single-molecule, real-time (SMRT) sequencing with mRNA quantitation using short-read mRNA sequencing to characterize changes in mRNA processing in the epithelial response to respiratory syncytial virus (RSV) replication. Mock or RSV-infected human small-airway epithelial cells (hSAECs) were profiled using SMRT sequencing and the curated transcriptome analyzed by structural and quality annotation of novel transcript isoforms (SQANTI). We identified 113,082 unique isoforms; 28,561 represented full splice matches, and 45% of genes expressed six or greater AS mRNA isoforms. Identification of differentially expressed AS isoforms was accomplished by mapping a short-read RNA sequencing expression matrix to the curated transcriptome, and 905 transcripts underwent differential polyadenylation site analysis enriched in protein secretion, translation, and mRNA degradation. We focused on 355 genes showing differential isoform utilization (DIU), indicating where a new AS isoform becomes a major fraction of mRNA isoforms expressed. In pathway and network enrichment analyses, we observed that DIU transcripts are substantially enriched in cell cycle control and IIR pathways. Interestingly, the RelA/IRF7 innate regulators showed substantial DIU where major transcripts included distinct isoforms with exon occlusion, intron inclusion, and alternative transcription start site utilization. We validated the presence of RelA and IRF7 AS isoforms as well as their induction by RSV using eight isoform-specific RT-PCR assays. These isoforms were identified in both immortalized and primary small-airway epithelial cells. We concluded that the cell cycle and IIR are differentially spliced in response to RSV. These data indicate that substantial post-transcriptional complexity regulates the antiviral response.


Subject(s)
Alternative Splicing , Immunity, Innate , RNA, Messenger/genetics , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/physiology , Epithelial Cells/immunology , Epithelial Cells/virology , Humans , RNA Processing, Post-Transcriptional , RNA, Messenger/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
12.
Mol Cell Biochem ; 476(5): 2029-2037, 2021 May.
Article in English | MEDLINE | ID: mdl-33512636

ABSTRACT

Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease that is mainly characterized as abnormal activation of B cells. It is reported that radical s-adenosyl methionine domain-containing 2 (RSAD2) is overexpressed in CD19+ B cells of pSS patients, but its role in pSS B cells remains unknown. Herein, RSAD2 expression was upregulated in CD19+ B cells of pSS patients and positively correlated with the expression of interleukin-10 (IL-10) in serum. After CD40L stimulation, knockdown of RSAD2 significantly attenuated cell viability, the production levels of immunoglobins and the expression of IL-10, while promoted cell apoptosis of pSS CD19+ B cells. Mechanistically, knockdown of RSAD2 negatively regulated nuclear factor kappa-b (NF-κb) signaling pathway. In addition, overexpression of p65 prominently alleviated the inhibitory effect of RSAD2 knockdown on proliferation, immunoglobin production and IL-10 expression in CD40L-induced CD19+ B cells. Our study indicated that silencing RSAD2 attenuated pSS B cell hyperactivity via suppressing NF-κb signaling pathway, which might provide a potential therapeutic target for pSS treatment.


Subject(s)
B-Lymphocytes/immunology , Proteins/immunology , Signal Transduction/immunology , Sjogren's Syndrome/immunology , Transcription Factor RelA/immunology , B-Lymphocytes/pathology , Gene Knockdown Techniques , Humans , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Signal Transduction/genetics , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Transcription Factor RelA/genetics
13.
Biol Pharm Bull ; 44(3): 350-356, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33390425

ABSTRACT

Inflammation has an important role in ischemia-reperfusion (I/R) injury. Artesunate (ART) has anti-microbial and anti-inflammatory pharmacological activities, and it is used for various types of serious malaria, including cerebral malaria. ART maintains a high concentration in the brain but little is known about the neuroprotective effect of ART against brain I/R injury. We studied the neuroprotection of ART against brain I/R injury and its underlying mechanism. In this study, rats were subjected to middle cerebral artery occlusion (MCAO) for 2 h. After 24 h of reperfusion, neurological deficits, cerebrum water content, infarct volume, hematoxylin-eosin (H&E)-staining, myeloperoxidase (MPO) activity, and proinflammatory cytokine levels were measured. Administration of 20, 40, 80, and 160 mg/kg ART intraperitoneally (i.p.) 10 min after MCAO significantly decreased brain water content and improved neurological deficits in a dose-dependent manner. An 80 mg/kg dosage was optimal. ART significantly reduced infarct volume, suppressed MPO activity and diminished the expressions of toll-like receptor (TLR)-4, MyD88, nuclear factor-κB (NF-κB), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in the area of the ischemic cortex. The neuroprotective action of ART against focal cerebral I/R injury might be due to the attenuation of inflammation through the TLR-4/NF-κB pathway.


Subject(s)
Artesunate/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Artesunate/pharmacology , Brain/drug effects , Brain/immunology , Brain/pathology , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Interleukin-6/immunology , Male , Neuroprotective Agents/pharmacology , Neutrophils/drug effects , Neutrophils/immunology , Rats, Sprague-Dawley , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Toll-Like Receptor 4/immunology , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/immunology
14.
Arthritis Rheumatol ; 73(3): 490-497, 2021 03.
Article in English | MEDLINE | ID: mdl-32969189

ABSTRACT

OBJECTIVE: Monogenic Behçet's disease (BD)-like conditions are increasingly recognized and to date have been found to predominantly involve loss-of-function variants in TNFAIP3. This study was undertaken to identify genetic and pathobiologic mechanisms associated with a BD-like mucocutaneous ulcerative syndrome and neuromyelitis optica (NMO) occurring in 3 generations of an Irish family (n = 5 cases and 5 familial controls). METHODS: Whole-exome sequencing was used to identify potential pathogenic variants in affected family members and determine segregation between affected and unaffected individuals. Relative v-rel reticuloendotheliosis viral oncogene homolog A (RELA) expression in peripheral blood mononuclear cells was compared by Western blotting. Human epithelial and RelA-/- mouse fibroblast experimental systems were used to determine the molecular impact of the RELA truncation in response to tumor necrosis factor (TNF). NF-κB signaling, transcriptional activation, apoptosis, and cytokine production were compared between wild-type and truncated RELA in experimental systems and patient samples. RESULTS: A heterozygous cytosine deletion at position c.1459 in RELA was detected in affected family members. This mutation resulted in a frameshift p.His487ThrfsTer7, producing a truncated protein disrupting 2 transactivation domains. The truncated RELA protein lacks a full transactivation domain. The RELA protein variants were expressed at equal levels in peripheral mononuclear cells. RelA-/- mouse embryonic fibroblasts (MEFs) expressing recombinant human RELAp.His487ThrfsTer7 were compared to those expressing wild-type RELA; however, there was no difference in RELA nuclear translocation. In RelA-/- MEFs, expression of RELAp.His487ThrfsTer7 resulted in a 1.98-fold higher ratio of cleaved caspase 3 to caspase 3 induced by TNF compared to wild-type RELA (P = 0.036). CONCLUSION: Our data indicate that RELA loss-of-function mutations cause BD-like autoinflammation and NMO via impaired NF-κB signaling and increased apoptosis.


Subject(s)
Apoptosis/genetics , Behcet Syndrome/genetics , Cytokines/immunology , NF-kappa B/immunology , Neuromyelitis Optica/genetics , Transcription Factor RelA/genetics , Adolescent , Adult , Animals , Apoptosis/immunology , Behcet Syndrome/immunology , Child , Female , Fibroblasts , Frameshift Mutation , Humans , Ireland , Loss of Function Mutation , Male , Mice , Mice, Knockout , Mice, Transgenic , Neuromyelitis Optica/immunology , Oral Ulcer/genetics , Oral Ulcer/immunology , Pedigree , Skin Ulcer/genetics , Skin Ulcer/immunology , Transcription Factor RelA/immunology , White People , Young Adult
15.
FEBS J ; 288(2): 640-662, 2021 01.
Article in English | MEDLINE | ID: mdl-32386462

ABSTRACT

Nuclear factor 'κ-light-chain-enhancer' of activated B cells (NF-κB) signaling is a signaling pathway used by most immune cells to promote immunostimulatory functions. Recent studies have indicated that regulatory T cells (Treg) differentially integrate TCR-derived signals, thereby maintaining their suppressive features. However, the role of NF-κB signaling in the activation of human peripheral blood (PB) Treg has not been fully elucidated so far. We show that the activity of the master transcription factor forkhead box protein 3 (FOXP3) attenuates p65 phosphorylation and nuclear translocation of the NF-κB proteins p50, p65, and c-Rel following activation in human Treg. Using pharmacological and genetic inhibition of canonical NF-κB signaling in FOXP3-transgenic T cells and PB Treg from healthy donors as well as Treg from a patient with a primary NFKB1 haploinsufficiency, we validate that Treg activation and suppressive capacity is independent of NF-κB signaling. Additionally, repression of residual NF-κB signaling in Treg further enhances interleukin-10 (IL-10) production. Blockade of NF-κB signaling can be exploited for the generation of in vitro induced Treg (iTreg) with enhanced suppressive capacity and functional stability. In this respect, dual blockade of mammalian target of rapamycin (mTOR) and NF-κB signaling was accompanied by enhanced expression of the transcription factors FOXP1 and FOXP3 and demethylation of the Treg-specific demethylated region compared to iTreg generated under mTOR blockade alone. Thus, we provide first insights into the role of NF-κB signaling in human Treg. These findings could lead to strategies for the selective manipulation of Treg and the generation of improved iTreg for cellular therapy.


Subject(s)
Forkhead Transcription Factors/immunology , Haploinsufficiency/immunology , NF-kappa B p50 Subunit/immunology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , Transcription Factor RelA/immunology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , Cell Nucleus/drug effects , Cell Nucleus/immunology , Cell Nucleus/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Lymphocyte Activation , NF-kappa B p50 Subunit/deficiency , NF-kappa B p50 Subunit/genetics , Phosphorylation/drug effects , Primary Cell Culture , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Thiazoles/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics
16.
J Agric Food Chem ; 68(42): 11747-11757, 2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33047600

ABSTRACT

Pelvic inflammatory disease (PID) is a common inflammation in the upper reproductive tract in women and may cause serious and costly consequences without effective treatment. Engeletin is a flavanonol glycoside and a naturally derived aldose reductase (AR) inhibitor that is widely distributed in vegetables, fruits, and plant-based foods. The present study investigated the anti-PID activity of engeletin in a mucilage-induced rat model of PID and LPS-stimulated RAW 264.7 macrophages. Engeletin significantly reduced inflammation and ameliorated the typical uterine pathological changes in PID rats. Engeletin also inhibited AR-dependent PLC/PKC/NF-κB and MAPK inflammatory pathways, as indicated by the suppression of the phosphorylation levels of PLC, PKC, p38, ERK, and JNK and the nuclear translocation of NF-κB p65. In vitro studies demonstrated that engeletin significantly inhibited inflammatory mediator expression and enhanced the phagocytic ability of LPS-induced RAW 264.7 macrophages. RNA interference of AR prevented the engeletin-induced inhibition of inflammatory mediators. Engeletin also inhibited AR-dependent PLC/PKC/NF-κB and MAPK inflammatory pathways, which was consistent with the in vivo results. These findings support engeletin as a potential agent for prevention or treatment of PID.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Anti-Inflammatory Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Flavonols/administration & dosage , Glycosides/administration & dosage , Pelvic Inflammatory Disease/diet therapy , Protein Kinase C/immunology , Transcription Factor RelA/immunology , Type C Phospholipases/immunology , Aldehyde Reductase/genetics , Aldehyde Reductase/immunology , Animals , Female , Humans , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/immunology , Mice , Pelvic Inflammatory Disease/genetics , Pelvic Inflammatory Disease/immunology , Protein Kinase C/genetics , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/genetics , Type C Phospholipases/genetics
17.
Vet Microbiol ; 251: 108892, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33126183

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) is an enveloped, single-stranded positive-sense RNA virus that belongs to a porcine entero-pathogenic alphacoronavirus, causing lethal watery diarrhea in piglets. Despite existing study reports the sole accessory protein ORF3 identified as NF-κB antagonist, the contribution of PEDV ORF3 to production of the pro-inflammatory cytokines mediated by NF-κB signaling remains largely unknown. Thus in this present study, we showed that PEDV ORF3 protein significantly inhibited the productions of pro-inflammatory cytokines interleukin-6 (IL-6) and IL-8. The phosphorylation of IκBα was inhibited by ORF3 protein, but no degradation of IκBα was induced in ORF3-expressing cells. Furthermore, PEDV ORF3 inhibited NF-κB activation through preventing nuclear factor p65 phosphorylation and down-regulating p65 expression level, as well as interfering nuclear translocation of p65, eventually resulting into the inhibition of IL-6 and IL-8 production. Our study definitely links PEDV ORF3 to inhibition of pro-inflammatory cytokines production, which will provide new insight into the molecular mechanisms of NF-κB activity inhibited by PEDV proteins to facilitate virus evasion of host innate immune.


Subject(s)
Interleukin-6/antagonists & inhibitors , Interleukin-8/antagonists & inhibitors , Porcine epidemic diarrhea virus/genetics , Transcription Factor RelA/genetics , Viral Proteins/genetics , Viral Proteins/immunology , Animals , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Interleukin-6/immunology , Interleukin-8/immunology , Porcine epidemic diarrhea virus/immunology , Signal Transduction , Swine , Transcription Factor RelA/immunology , Vero Cells , Virus Replication
18.
J Leukoc Biol ; 108(6): 1747-1764, 2020 12.
Article in English | MEDLINE | ID: mdl-32991757

ABSTRACT

Tuberculosis is a serious public health problem aggravated by the slow progress in the development of new anti-tuberculosis drugs. The hyper-reactive TB patients have suffered from chronic inflammation which could cause deleterious effects on their bodies. Therefore, it is imperative to develop an adjunctive therapy based on inflammatory modulation during Mycobacterium tuberculosis (Mtb) infection. The present study aims to investigate the immune regulatory effects of Andrographolide (Andro) on Mtb-infected macrophages and its underlying mechanisms. The results showed that Andro inhibits the production of IL-1ß and other inflammatory cytokines in a dose-dependent manner. The down-regulation of IL-1ß expression causes the declining expression of IL-8 and MCP-1 in lung epithelial cells which were co-cultured with Mtb-infected macrophages.  The inhibition of the activation of NF-κB pathway, but not the inhibition of MAPK signaling pathway, accounts for the anti-inflammatory role of Andro. Further studies elucidated that Andro could evoke the activation of autophagy to degrade NLRP3, which ultimately inhibited inflammasome activation and subsequent IL-1ß production. Finally, the relevant results demonstrated that Andro inhibited the Notch1 pathway to down-regulate the phosphorylation of Akt/mTOR and NF-κB p65 subunit. Taken together, Andro has been found to suppress the Notch1/Akt/NF-κB signaling pathway. Both Akt inhibition-induced autophagy and inhibition of the NF-κB pathway contributed to restraining the activation of NLRP3 inflammasome and subsequent IL-1ß production. Then, the decreased production of IL-1ß influenced chemokine expression in lung epithelial cells. Based on these results, anti-inflammatory effect of Andro in TB infection is merit further investigation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Diterpenes/pharmacology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Proto-Oncogene Proteins c-akt/immunology , Receptor, Notch1/immunology , Transcription Factor RelA/immunology , Tuberculosis/immunology , Animals , Female , Macrophages/pathology , Mice , Tuberculosis/drug therapy , Tuberculosis/pathology
19.
Commun Biol ; 3(1): 515, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948804

ABSTRACT

Regulating the amount of proteins in living cells is a powerful approach for understanding the functions of the proteins. Immunomodulatory drugs (IMiDs) induce the degradation of neosubstrates by interacting with celebron (CRBN) in the cullin E3 ubiquitin ligase complex (CRL4CRBN). Here, we developed the IMiD-dependent Sal-like protein 4 (SALL4) degron (S4D) system for chemical protein knockdown. In transient assays, an N- or C-terminal S4D tag induced the degradation of proteins localized to various subcellular compartments, including the plasma membrane. The activity of luciferase-S4D was reduced by 90% within 3 h of IMiD treatment. IMiD treatment reduced the expression of endogenous S4D-fused RelA and IκBα in knock-in (KI) experiments. Interestingly, the IκBα knockdown suggested that there may be another, unknown mechanism for RelA translocation to the nucleus. Furthermore, 5-hydroxythalidomide as a thalidomide metabolite specifically degradated S4D-tagged protein. These results indicate that the S4D system is a useful tool for cellular biology.


Subject(s)
Immunologic Factors/genetics , Proteolysis , Thalidomide/metabolism , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Cell Membrane/genetics , Cell Membrane/immunology , Gene Knockdown Techniques/methods , HeLa Cells , Humans , Immunologic Factors/immunology , Immunologic Factors/pharmacology , Substrate Specificity , Thalidomide/analogs & derivatives , Thalidomide/immunology , Thalidomide/pharmacology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription Factors/immunology , Transcriptional Elongation Factors/genetics , Ubiquitin-Protein Ligases/immunology
20.
Int Immunopharmacol ; 88: 106923, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32889238

ABSTRACT

Protein arginine deiminase 4 (PAD4) serves a critical role in differentiation, development and apoptosis through gene regulation and has emerged as a potential therapeutic target for the treatment of various diseases. However, the roles of PAD4 in lipopolysaccharide (LPS)-induced acute lung injury (ALI) remain largely unknown. To investigate the roles of PAD4 during LPS-induced ALI, the present study detected the trend of PAD4 expression in the lung tissues of ALI mice. Subsequently, the efficiency of TDFA on PAD4 and citrullinated H3 histone were detected. And then, histology, the wet/dry weight ratio, survival rate, activated cells infiltration, oxidative stress levels, tight junction proteins and proinflammatory cytokine expression were detected. In addition, the level of transepithelial electrical resistance (TEER) was assessed. Finally, the level of nuclear P65, total phosphorylated P65 and P65 were measured in vivo and in vitro. The results showed that PAD4 expression was upregulated in the lung tissues of LPS-induced ALI. TDFA efficiently decreased the severity of the lung edema, attenuated the severity of pulmonary injury and improved the survival rate following lethal LPS administration. Besides, TDFA reduced activated cells infiltration and suppressed inflammation related parameters, including proinflammatory cytokines production (TNF-α, IL-6 and IL-1ß) and oxidative stress (MDA, GSH and SOD). Furthermore, TDFA reversed the TEER downregulation tendency and tight junction proteins (ZO-1, Occludin, Claudin-4) levels that represent the integrity of alveolar epithelium. Eventually, TDFA exerts its protective roles through modulating nuclear localization of transcription factor NF-κB P65 in epithelial cells. Taken together, these results indicate that PAD4 inhibition may serve as a promising therapeutic approach for LPS-induced ALI.


Subject(s)
Acute Lung Injury/drug therapy , Amidines/therapeutic use , Epithelial Cells/drug effects , Protective Agents/therapeutic use , Protein-Arginine Deiminase Type 4/antagonists & inhibitors , Transcription Factor RelA/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Amidines/pharmacology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Cytokines/immunology , Epithelial Cells/immunology , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Male , Mice, Inbred C57BL , Protective Agents/pharmacology , Protein-Arginine Deiminase Type 4/immunology , Tight Junction Proteins/genetics , Tight Junction Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...