Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.181
Filter
1.
STAR Protoc ; 5(1): 102873, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38427566

ABSTRACT

Here, we present a protocol to deliver nanoliter volumes of Toll-like receptor (TLR) agonist onto a culture of nuclear factor κB (NF-κB) reporter macrophages using fluidic force microscopy and a micron-scale probe. We describe steps for quantifying the dose of agonist by modeling their diffusion with experimental inputs. We then detail procedures for quantifying and categorizing macrophage responses to individual and varied doses and combining agonist concentration and macrophage response to analyze the NF-κB response to localized TLR stimulation. For complete details on the use and execution of this protocol, please refer to Mulder et al. (2024).1.


Subject(s)
NF-kappa B , Toll-Like Receptors , NF-kappa B/physiology , Microscopy, Atomic Force , Toll-Like Receptor 4 , Macrophages
2.
Sci Rep ; 14(1): 5078, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38429394

ABSTRACT

Ferroptosis is a recently identified form of programmed cell death that plays an important role in the pathophysiological process of osteoarthritis (OA). Herein, we investigated the protective effect of moderate mechanical stress on chondrocyte ferroptosis and further revealed the internal molecular mechanism. Intra-articular injection of sodium iodoacetate (MIA) was conducted to induce the rat model of OA in vivo, meanwhile, interleukin-1 beta (IL-1ß) was treated to chondrocytes to induce the OA cell model in vitro. The OA phenotype was analyzed by histology and microcomputed tomography, the ferroptosis was analyzed by transmission electron microscope and immunofluorescence. The expression of ferroptosis and cartilage metabolism-related factors was analyzed by immunohistochemical and Western blot. Animal experiments revealed that moderate-intensity treadmill exercise could effectively reduce chondrocyte ferroptosis and cartilage matrix degradation in MIA-induced OA rats. Cell experiments showed that 4-h cyclic tensile strain intervention could activate Nrf2 and inhibit the NF-κB signaling pathway, increase the expression of Col2a1, GPX4, and SLC7A11, decrease the expression of MMP13 and P53, thereby restraining IL-1ß-induced chondrocyte ferroptosis and degeneration. Inhibition of NF-κB signaling pathway relieved the chondrocyte ferroptosis and degeneration. Meanwhile, overexpression of NF-κB by recombinant lentivirus reversed the positive effect of CTS on chondrocytes. Moderate mechanical stress could activate the Nrf2 antioxidant system, inhibit the NF-κB p65 signaling pathway, and inhibit chondrocyte ferroptosis and cartilage matrix degradation by regulating P53, SLC7A11, and GPX4.


Subject(s)
Ferroptosis , Osteoarthritis , Stress, Mechanical , Animals , Rats , Chondrocytes/metabolism , Interleukin-1beta/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , NF-kappa B/physiology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , X-Ray Microtomography , Transcription Factor RelA/metabolism , Transcription Factor RelA/physiology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology
3.
Biomed Pharmacother ; 156: 113951, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36411636

ABSTRACT

Polyphenol compounds are natural antioxidants, which are rich in anti-inflammatory and antioxidant components. They have a wide range of medicinal benefits that are believed to improve human health across various aspects; especially its anticancer effect has been gradually confirmed. The anticancer effect of polyphenols is mainly based on their strong antioxidant and immunomodulatory effects. The innate and adaptive immune responses as well as the development and maintenance of cells and tissues of the immune system are regulated by the NF-κB family of transcription factors. Dysregulation of NF-κB can lead to autoimmune diseases, chronic inflammation, and even cancer. Polyphenol compounds can exert antioxidant and immunomodulatory effects by targeting NF-κB, thus hindering the occurrence and development of tumors.Polyphenol compounds have unique advantages over conventional anticancer therapies such as chemotherapy because they have few side effects and do not cause toxicity to healthy cells. Additionally, they can attenuate the toxic effects of current anticancer therapies. Based on these characteristics, polyphenols have great potential in the prevention and treatment of cancer. This article systematically summarizes the mechanism of NF-κB in tumor genesis, progression, metastasis, angiogenesis, and drug resistance. In addition, we present the anticancer effect of polyphenol compounds by targeting NF-κB during the different stages of tumorigenesis.


Subject(s)
NF-kappa B , Neoplasms , Humans , NF-kappa B/physiology , Polyphenols/pharmacology , Polyphenols/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , I-kappa B Proteins , Neoplasms/drug therapy
4.
Life Sci ; 307: 120860, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35940220

ABSTRACT

Amidst several pathophysiological cascades, Advanced Glycation End products (AGEs) have been identified as a pivotal aetiology behind the pathogenesis and progression of cardiovascular disorders, by inducing oxidative stress and inflammation of myocardial and vascular tissues. Non-enzymatic glycation of reducing sugars with amino acids in proteins, lipids, and nucleic acids produce AGEs, which are a diverse set of compounds. Although AGEs are mostly generated endogenously, current research suggests that nutrition is a major exogenous source of AGEs. Extracellular and intracellular structure and function are affected by the presence and accumulation of AGEs in several cardiac cell types. AGEs give rise to several microvascular and macrovascular problems by establishing cross-links between molecules in the extracellular matrix's basement membrane as well as interacting with receptors for advanced glycation end products (RAGE). The transcription factor nuclear factor kappa B and its RAGE target genes are upregulated when RAGE is activated by AGEs. Engagement increases oxidative stress and triggers inflammatory and fibrotic responses, all of which contribute to the onset and progression of life-threatening cardiovascular diseases. This article discusses the probable targets of glycation in cardiac cells, as well as the underlying mechanisms that lead to heart failure.


Subject(s)
Cardiovascular Diseases , Nucleic Acids , Amino Acids , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Glycation End Products, Advanced/metabolism , Humans , Lipids , NF-kappa B/physiology , Receptor for Advanced Glycation End Products/metabolism , Sugars
5.
Inflamm Res ; 71(2): 205-214, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35064305

ABSTRACT

OBJECTIVE: This study aimed to investigate the relationship between miR-21 and lipopolysaccharide (LPS)-induced myocardial injury and its molecular and regulatory mechanisms. METHODS: We constructed LPS-mediated myocardial injury model using C57BL/6J mice and H9c2 cells. In-vivo, in-vitro, RIP and dual-luciferase reporter assays were used to determine the effect of miR-21 on myocardial injury. RESULTS: In-vivo and in-vitro results showed that the expression of miR-21 was increased in LPS-treated H9c2 cells and myocardial tissues of mice, and the pro-inflammatory cytokines (IL-1ß, IL-6, IL-8 and TNF-α) and NF-κB pathway were activated in LPS-treated H9c2 cells. Besides, the B-cell lymphoma-2 (Bcl-2) and cyclin-dependent kinase 6 (CDK6) expression levels decreased, while Bax and cleaved caspase 9 levels increased in LPS-treated H9c2 cells. Inhibition of miR-21 could suppress LPS-induced apoptosis, inflammatory reactions and NF-κB activation to attenuate LPS-induced myocardial injury in H9c2 cells, and effectively improve survival of mice with sepsis. Most importantly, Bcl-2 and CDK6 were found to be the direct target of miR-21 using dual-luciferase reporter and RNA immunoprecipitation assays. Further gain-of-function assay demonstrated that Bcl-2 or CDK6 over-expression promoted the protective effects of miR-21 inhibitor on LPS-mediated myocardial cells. CONCLUSION: Our findings revealed that the down-regulation or antagonism of miR-21 protects myocardial cells against LPS-induced apoptosis and inflammation through up-regulating Bcl-2 and CDK6 expression, which provided a new insight for prevention and treatment of myocardial injury.


Subject(s)
Cyclin-Dependent Kinase 6/genetics , Heart/drug effects , MicroRNAs/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis/drug effects , Cells, Cultured , Heart/physiology , Inflammation/prevention & control , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-kappa B/physiology
6.
Inflamm Res ; 71(2): 227-241, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34993560

ABSTRACT

OBJECTIVE: Trimetazidine (TMZ) exerts a strong inhibitory effect on ischemia/reperfusion (I/R) injury. Inflammation plays a key role in I/R injury. We hypothesized that TMZ may protect cardiomyocytes from I/R injury by inhibiting inflammation. METHODS: The left anterior descending coronary artery was ligated for 30 min followed by 6 h of reperfusion to establish a model of I/R injury. H9c2 cardiomyocytes were subjected to 2 h of hypoxia and 3 h of normoxic conditions to establish a model of hypoxia/reoxygenation (H/R) injury. We monitored the change in pyroptosis by performing Western blot analysis, microscopy and ELISA. RESULTS: I/R and H/R treatment stimulated gasdermin D-N domain (GSDMD-N) expression in cardiomyocytes (sham onefold vs. I/R 2.5-fold; control onefold vs. H/R 2.0-fold). Moreover, TMZ increased the viability of H9c2 cardiomyocytes subjected to H/R treatment (H/R 65.0% vs. H/R + TMZ 85.3%) and reduced the infarct size in vivo (I/R 47.0% vs. I/R + TMZ 28.3%). H/R and I/R treatment increased the levels of TLR4, MyD88, phospho-NF-κB p65 and the NLRP3 inflammasome; however, TMZ reduced the expression of these proteins. Additionally, TMZ inhibited noncanonical inflammasome signaling induced by I/R injury. CONCLUSIONS: In summary, TMZ alleviated pyroptosis induced by myocardial I/R injury through the TLR4/MyD88/NF-κB/NLRP3 inflammasome pathway. Therefore, TMZ represents an alternative treatment for myocardial I/R injury.


Subject(s)
Myocardial Reperfusion Injury/drug therapy , Phosphate-Binding Proteins/antagonists & inhibitors , Pore Forming Cytotoxic Proteins/antagonists & inhibitors , Pyroptosis/drug effects , Trimetazidine/pharmacology , Animals , Male , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-kappa B/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Phosphate-Binding Proteins/physiology , Pore Forming Cytotoxic Proteins/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Toll-Like Receptor 4/physiology
7.
Nephron ; 146(1): 84-98, 2022.
Article in English | MEDLINE | ID: mdl-34518457

ABSTRACT

BACKGROUND: Glomerular endothelial cell damage plays an important role in the occurrence and development of diabetic nephropathy (DN). OBJECTIVES: This study aimed to clarify the role of XCL1 in DN-mediated glomerular endothelial cell apoptosis and whether the function was related to the activation of the p53/nuclear factor-kappa B (NF-κB) signaling pathway. METHODS: Candidate biomarkers were identified by least absolute shrinkage and selection operator (LASSO) regression model analysis. The area under the receiver operating characteristic curve value was calculated and used to evaluate the discriminating ability. Cell viability, apoptosis, and interleukin-1ß and tumor necrosis factor-α expression at messenger RNA and protein levels were detected by using the Cell Counting Kit-8, flow cytometry, ELISA, real-time polymerase chain reaction, and Western blotting assays. In vivo studies were conducted in the DN mice. RESULTS: The LASSO regression model displayed good discriminating performance, with a C-index of 0.803 and good calibration, and high XCL1 expression was identified as the predicting factor for DN in diabetes mellitus patients. XCL1 expression was upregulated in glomeruli of db/db mice, which was closely related to the expression of its receptor (XCR1). XCL1 overexpression played an important role in the apoptosis and inflammatory response of high glucose (HG)-treated human renal glomerular endothelial cells. Meanwhile, the expression of p53 and the levels of inflammatory cytokines were upregulated upon XCL1 overexpression. p53 silencing with its inhibitor blocked the apoptotic response and inflammatory response in XCL1-overexpressed cells exposed to HG. Besides, the XCL1 overexpression-induced downregulation of NF-κB was reversed by pifithrin-α pretreatment. CONCLUSIONS: Our findings in this work provided the mechanistic insights into the effects of XCL1 on the modulation of DN development, illustrating that XCL1 might serve as an essential prognostic indicator and therapeutic target for DN progression.


Subject(s)
Apoptosis/physiology , Chemokines, C/physiology , Diabetic Nephropathies/physiopathology , Inflammation/physiopathology , Kidney Glomerulus/pathology , NF-kappa B/physiology , Tumor Suppressor Protein p53/physiology , Adult , Aged , Animals , Disease Progression , Endothelial Cells/pathology , Female , Humans , Male , Mice , Middle Aged
8.
Sci China Life Sci ; 65(3): 588-603, 2022 03.
Article in English | MEDLINE | ID: mdl-34125371

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease that eventually leads to disability. Inflammatory cell infiltration, severe joint breaking and systemic bone loss are the main clinical symptoms. In this study, we established a collagen-induced arthritis (CIA) model and found a large number of M1 macrophages and pyroptosis, which are important sources of proinflammatory cytokines. Punicalagin (PUN) is an active substance extracted from pomegranate peel. We found that it inhibited joint inflammation, cartilage damage and systemic bone destruction in CIA mice. PUN effectively alleviated the high expression of inflammatory cytokines in synovial tissue in vivo. PUN treatment shifted macrophages from the M1 phenotype to the M2 phenotype after stimulation with lipopolysaccharide (LPS) and interferon (IFN)-γ. The expression of inducible nitric oxide synthase (iNOS) and other proinflammatory cytokines released by M1 macrophages was decreased in the PUN treatment group. However, simultaneously, the expression of markers of anti-inflammatory M2 macrophages, such as arginase (Arg)-1 and interleukin (IL)-10, was increased. In addition, PUN treatment attenuated pyroptosis by downregulating the expression of NLRP3 and caspase-1, thereby preventing inflammatory cell death resulting from the release of IL-1ß and IL-18. Mechanistically, PUN inhibited the activation of receptor activators of the nuclear factor-κB (NF-κB) signaling pathway, which contributes to M1 polarization and pyroptosis of macrophages. We concluded that PUN ameliorated pathological inflammation by inhibiting M1 phenotype polarization and pyroptosis and has great potential as a therapeutic treatment for human RA.


Subject(s)
Arthritis, Experimental/drug therapy , Hydrolyzable Tannins/pharmacology , Macrophages/drug effects , NF-kappa B/physiology , Pyroptosis/drug effects , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Caspase 1/physiology , Cells, Cultured , Cytokines/analysis , Down-Regulation , Hydrolyzable Tannins/therapeutic use , Male , Mice , Mice, Inbred DBA , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
9.
Clin Immunol ; 234: 108914, 2022 01.
Article in English | MEDLINE | ID: mdl-34954131

ABSTRACT

Increasing fine particulate matter (PM2.5) and epigenetic modifications are closely associated with the pathogenesis of asthma, but the definite mechanism remains unclear. The traffic-related PM2.5 exposure aggravated pulmonary inflammation and changed the methylation level of interferon gamma (Ifng) and interleukin (Il)4 genes, and then altered levels of affiliated cytokines of IFN-γ and IL-4 in rats with allergic airway inflammation. It also increased the level of miR146a and decreased the level of miR31. In addition, transcription factors of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 6 (Stat6) rose; forkhead box P3 (Foxp3) and signal transducer and activator of transcription 4 (Stat4) lowered. The traffic-related PM2.5 altered epigenetic modifications in allergic airway inflammation of rats leading to inflammation exacerbation through impaired regulatory T (Treg) cells function and T-helper type 1 (Th1)/Th2 cells imbalance, which provided a new target for the treatment and control of asthma.


Subject(s)
Asthma/etiology , DNA Methylation , Particulate Matter/toxicity , Vehicle Emissions/toxicity , Animals , Asthma/genetics , Asthma/immunology , Cytokines/analysis , Forkhead Transcription Factors/physiology , Interferon-gamma/genetics , Interferon-gamma/physiology , Interleukin-4/genetics , Interleukin-4/physiology , Male , MicroRNAs/analysis , NF-kappa B/physiology , Rats , Rats, Sprague-Dawley , STAT Transcription Factors/analysis , T-Lymphocytes, Regulatory/immunology
10.
Mediators Inflamm ; 2021: 7042148, 2021.
Article in English | MEDLINE | ID: mdl-34858081

ABSTRACT

Endothelial inflammation is a crucial event in the initiation of atherosclerosis. Here, we identify Ataxin-10 protein as a novel negative modulator of endothelial activation by suppressing IRF-1 transcription activity. The protein level of Ataxin-10 is relatively higher in human vascular endothelial cells, which can be significantly suppressed by TNF-α in both HUVECs and HLMECs. Overexpression of Ataxin-10 markedly inhibited the mRNA expressions of VCAM-1 and several cytokines including MCP-1, CXCL-1, CCL-5, and TNF-α; thus, it can also suppress monocyte adhesion to endothelial cells. Accordingly, Ataxin-10 silencing promoted endothelial inflammation. However, Ataxin-10 did not affect the MAPK/NF-κB signaling pathway stimulated by TNF-α in HUVECs. Using the yeast two-hybrid assay, we found that Ataxin-10 can directly bind to interferon regulatory factor-1 (IRF-1). Upon TNF-α stimulation, Ataxin-10 promoted the cytoplasmic localization of IRF-1, which inhibited the transcription of VCAM-1. Moreover, knockdown of IRF-1 can eliminate the effect of Ataxin-10 on the expression of VCAM-1 in HUVECs induced by TNF-α. Taken together, these results indicate that Ataxin-10 inhibits endothelial cell activation and may serve as a promising therapeutic target for some vascular inflammatory-related diseases such as atherosclerosis.


Subject(s)
Ataxin-10/physiology , Endothelial Cells/drug effects , Inflammation/prevention & control , Interferon Regulatory Factor-1/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Atherosclerosis/etiology , Cells, Cultured , Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells , Humans , Monocytes/physiology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/analysis , Vascular Cell Adhesion Molecule-1/genetics
11.
Front Immunol ; 12: 763334, 2021.
Article in English | MEDLINE | ID: mdl-34950140

ABSTRACT

Periodontitis (PD) is a common chronic infectious disease. The local inflammatory response in the host may cause the destruction of supporting periodontal tissue. Macrophages play a variety of roles in PD, including regulatory and phagocytosis. Moreover, under the induction of different factors, macrophages polarize and form different functional phenotypes. Among them, M1-type macrophages with proinflammatory functions and M2-type macrophages with anti-inflammatory functions are the most representative, and both of them can regulate the tendency of the immune system to exert proinflammatory or anti-inflammatory functions. M1 and M2 macrophages are involved in the destructive and reparative stages of PD. Due to the complex microenvironment of PD, the dynamic development of PD, and various local mediators, increasing attention has been given to the study of macrophage polarization in PD. This review summarizes the role of macrophage polarization in the development of PD and its research progress.


Subject(s)
Macrophages/physiology , Periodontitis/immunology , Animals , Cell Polarity , Cytokines/physiology , Humans , Janus Kinases/physiology , NF-kappa B/physiology , Periodontitis/drug therapy , Periodontitis/etiology , Periodontium/immunology , STAT Transcription Factors/physiology , Signal Transduction/physiology
12.
Elife ; 102021 12 06.
Article in English | MEDLINE | ID: mdl-34866574

ABSTRACT

The pathogenesis of COVID-19 is associated with a hyperinflammatory response; however, the precise mechanism of SARS-CoV-2-induced inflammation is poorly understood. Here, we investigated direct inflammatory functions of major structural proteins of SARS-CoV-2. We observed that spike (S) protein potently induced inflammatory cytokines and chemokines, including IL-6, IL-1ß, TNFα, CXCL1, CXCL2, and CCL2, but not IFNs in human and mouse macrophages. No such inflammatory response was observed in response to membrane (M), envelope (E), and nucleocapsid (N) proteins. When stimulated with extracellular S protein, human and mouse lung epithelial cells also produced inflammatory cytokines and chemokines. Interestingly, epithelial cells expressing S protein intracellularly were non-inflammatory, but elicited an inflammatory response in macrophages when co-cultured. Biochemical studies revealed that S protein triggers inflammation via activation of the NF-κB pathway in a MyD88-dependent manner. Further, such an activation of the NF-κB pathway was abrogated in Tlr2-deficient macrophages. Consistently, administration of S protein-induced IL-6, TNF-α, and IL-1ß in wild-type, but not Tlr2-deficient mice. Notably, upon recognition of S protein, TLR2 dimerizes with TLR1 or TLR6 to activate the NF-κB pathway. Taken together, these data reveal a mechanism for the cytokine storm during SARS-CoV-2 infection and suggest that TLR2 could be a potential therapeutic target for COVID-19.


Subject(s)
Inflammation/virology , NF-kappa B/physiology , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/immunology , Toll-Like Receptor 2/genetics , A549 Cells , Animals , HEK293 Cells , Humans , Mice , Signal Transduction , Toll-Like Receptor 2/metabolism
13.
Mol Pharm ; 18(12): 4322-4330, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34734526

ABSTRACT

Parathyroid hormone-related protein (PTHrP), which is secreted from a tumor, contributes to the progression of cachexia, a condition that is observed in half of all cancer patients. Although drug clearance was reported to decrease in patients with cancer cachexia, the details have not been clarified. The present study reports on an investigation of whether PTHrP is involved in the alternation of drug metabolism in cases of cancer cachexia. Cancer cachexia model rats with elevated serum PTHrP levels showed a significant decrease in hepatic and intestinal CYP3A2 protein expression. When midazolam, a CYP3A substrate drug, was administered intravenously or orally to the cancer cachexia rats, its area under the curve (AUC) was increased by about 2 and 5 times, as compared to the control group. Accordingly, the bioavailability of midazolam was increased by about 3 times, thus enhancing its pharmacological effect. In vitro experiments using HepG2 cells and Caco-2 cells showed that the addition of serum from cancer cachexia rats or active PTHrP (1-34) to each cell resulted in a significant decrease in the expression of CYP3A4 mRNA. Treatment with a cell-permeable cAMP analog also resulted in a decreased CYP3A4 expression. Pretreatment with protein kinase A (PKA), protein kinase C (PKC), and nuclear factor-kappa B (NF-κB) inhibitors recovered the decrease in CYP3A4 expression that was induced by PTHrP (1-34). These results suggest that PTHrP suppresses CYP3A expression via the cAMP/PKA/PKC/NF-κB pathway. Therefore, it is likely that PTHrP would be involved in the changes in drug metabolism observed in cancer cachexia.


Subject(s)
Cachexia/metabolism , Cytochrome P-450 CYP3A/genetics , Neoplasms/complications , Parathyroid Hormone-Related Protein/physiology , Animals , Caco-2 Cells , Hep G2 Cells , Humans , Liver/enzymology , Male , Midazolam/pharmacokinetics , NF-kappa B/physiology , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley
14.
J Nat Prod ; 84(11): 2808-2821, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34726063

ABSTRACT

Chromatographic fractionation of Sigesbeckia glabrescens led to the identification of 10 new sesquiterpene lactones, named siegesbeckialides I-O (1-7) and glabrescones A-C (8-10), along with 14 known analogues. An anti-inflammatory activity assay showed that siegesbeckialide I (1) most potently inhibited LPS-induced NO production in RAW264.7 murine macrophages. Furthermore, siegesbeckialide I suppressed the protein expression of iNOS and COX2, as well as the release of PGE2, IL-1ß, IL-6, and TNF-α in LPS-stimulated RAW264.7 cells. Mechanistically, siegesbeckialide I directly binds to inhibitors of IKKα/ß and suppresses their phosphorylation. This leads to the inhibition of IKKα/ß-mediated phosphorylation and degradation of inhibitor α of NF-κB (IκBα), as well as the activation of NF-κB signaling.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Asteraceae/chemistry , I-kappa B Kinase/antagonists & inhibitors , Lactones/pharmacology , Sesquiterpenes/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , HEK293 Cells , Humans , Lactones/chemistry , Lactones/isolation & purification , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/physiology , Phosphorylation , RAW 264.7 Cells , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification
15.
Mediators Inflamm ; 2021: 6259381, 2021.
Article in English | MEDLINE | ID: mdl-34675753

ABSTRACT

OBJECTIVE: To determine the role of sodium butyrate in intestinal inflammation via regulation of high-mobility group box-1 (HMGB1), we analyzed the potential mechanism in necrotizing enterocolitis (NEC) in a neonatal mouse model. METHODS: A NEC model was created with hypoxia and cold exposure and artificial overfeeding. C57BL/6 neonatal mice were randomized into three groups: the control, untreated NEC, and sodium butyrate (150 mM)-pretreated NEC groups. Pathological variations in ileocecal intestinal tissue were observed by HE staining and scored in a double-blind manner. The mRNA expression levels of HMGB1, Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), and inflammatory cytokines in intestinal tissues were determined by quantitative real-time PCR. The protein levels of HMGB1 and associated cytokines in intestinal tissues were evaluated using ELISA. The relative protein expression levels of TLR4 and NF-κB in intestinal tissues were quantified by western blot. RESULTS: Sodium butyrate administration improved the body weight and survival rate of NEC mice; relieved intestinal pathological injury; reduced the intestinal expression of HMGB1, TLR4, NF-κB, interleukin- (IL-) 1ß, IL-6, IL-8, and TNF-α; and increased the intestinal expression of IL-10 (P < 0.05). Treatment with butyrate decreased the proportion of opportunistic Clostridium_sensu_stricto_1 and Enterococcus and increased the proportion of beneficial Firmicutes and Lactobacillus in the NEC model. CONCLUSIONS: Sodium butyrate intervention relieves intestinal inflammation and partially corrects the disrupted intestinal flora in mice with NEC.


Subject(s)
Butyric Acid/therapeutic use , Enterocolitis, Necrotizing/drug therapy , Animals , Butyric Acid/pharmacology , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/pathology , Female , HMGB1 Protein/genetics , HMGB1 Protein/physiology , Intestines/microbiology , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/physiology , Random Allocation , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
16.
Placenta ; 115: 97-105, 2021 11.
Article in English | MEDLINE | ID: mdl-34598084

ABSTRACT

INTRODUCTION: Excessive activation of maternal systemic inflammation is one of the underlying causes of pathology during the disease course of preeclampsia (PE). The triggering receptor expressed on myeloid cells-1 (TREM-1) participates in the development and persistence of inflammation. We hypothesized that dysregulated TREM-1 may be involved in the pathogenesis of PE by promoting the secretion of trophoblastic pro-inflammatory cytokines that augment inflammation. METHODS: The localization of TREM-1 in placenta and the extravillous trophoblast cell line (TEV-1) was determined by immunohistochemical staining. The expression level of TREM-1 and pro-inflammatory cytokines in placentas were compared between normal pregnancies and PE. We used lipopolysaccharide (LPS) to simulate trophoblastic inflammation. TEV-1 cells were transfected with TREM-1 plasmid and si-TREM-1 respectively, and then were incubated with LPS. The expression levels of pro-inflammatory cytokines and key molecules featured in nuclear transcription factor-kappaB (NF-κB) pathway were detected. Transwell assays were used to detect the effects of TREM-1 on cell migration and invasion. RESULTS: TREM-1 was localized on both villous trophoblasts (VTs) and extravillous trophoblasts (EVTs). TREM-1 and pro-inflammatory cytokines were up-regulated in preeclamptic placenta. Overexpression of TREM-1 promoted the activation of NF-κB pathway and the release of pro-inflammatory factors induced by LPS, and enhanced migration and invasion of TEV-1 cells. Inhibition of TREM-1 significantly attenuated LPS-induced effects and suppressed migration and invasion. DISCUSSION: This study suggested that TREM-1 was up-regulated in PE, and may promote the production of downstream inflammatory factors by activating NF-κB pathway in trophoblastic cells, thus exerting pro-inflammatory effects in the pathogenesis of PE.


Subject(s)
Inflammation/physiopathology , NF-kappa B/physiology , Pre-Eclampsia/physiopathology , Triggering Receptor Expressed on Myeloid Cells-1/physiology , Trophoblasts/physiology , Adult , Cell Line, Transformed , Female , Humans , Interleukins/genetics , Lipopolysaccharides/pharmacology , Placenta/chemistry , Pregnancy , RNA, Messenger/analysis , Transfection , Triggering Receptor Expressed on Myeloid Cells-1/analysis , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Trophoblasts/chemistry , Trophoblasts/drug effects , Tumor Necrosis Factor-alpha/genetics
17.
Int J Oncol ; 59(5)2021 11.
Article in English | MEDLINE | ID: mdl-34558639

ABSTRACT

The expression of microRNA­383 (miR­383) is downregulated in a variety of tumor tissues, and it exhibits antiproliferative activity in non­small cell lung cancer cells. In the present study, an association between the downregulation of miR­383 expression and the deletion of chr8p22 in patients with lung adenocarcinoma was identified. The promoting effect of miR­383 on cisplatin sensitivity was verified both in vivo and in vitro. Additionally, it was revealed that the expression of RNA binding motif protein 24 (RBM24) protein was regulated by and negatively correlated with miR­383 expression. Ectopic expression of RBM24 or inhibition of miR­383 decreased the chemosensitivity of parental A549 cells, whereas knockdown of RBM24 in cisplatin­resistant A549 cells increased chemosensitivity. Mechanistically, miR­383 interfered with the activation of nuclear factor κB (NF­κB) signaling through repression of RBM24­mediated phosphorylation of Rel­like domain­containing protein A and inhibitor α of NF­κB. Taken together, the downregulation of miR­383 induced RBM24 expression, which was mediated through the activation of NF­κB signaling, to contribute to chemotherapy resistance in lung adenocarcinoma cells. The results of the present study highlight potential therapeutic targets for the clinical reversal of the chemotherapy resistance in lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Cisplatin/pharmacology , Lung Neoplasms/drug therapy , MicroRNAs/physiology , NF-kappa B/physiology , RNA-Binding Proteins/physiology , Adenocarcinoma of Lung/mortality , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/mortality , Male , Mice , Mice, Inbred BALB C , Middle Aged , Prognosis , Signal Transduction/drug effects
18.
Int Immunopharmacol ; 100: 108071, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34482267

ABSTRACT

COVID-19 is the cause of a pandemic associated with substantial morbidity and mortality. As yet, there is no available approved drug to eradicate the virus. In this review article, we present an alternative study area that may contribute to the development of therapeutic targets for COVID-19. Growing evidence is revealing further pathophysiological mechanisms of COVID-19 related to the disregulation of inflammation pathways that seem to play a critical role toward COVID-19 complications. The NF-kB and JAK/STAT signaling pathways are highly activated in acute inflammation, and the excessive activity of these pathways in COVID-19 patients likely exacerbates the inflammatory responses of the host. A group of non-coding RNAs (miRNAs) manage certain features of the inflammatory process. In this study, we discuss recent advances in our understanding of miRNAs and their connection to inflammatory responses. Additionally, we consider the link between perturbations in miRNA levels and the onset of COVID-19 disease. Furthermore, previous studies published in the online databases, namely web of science, MEDLINE (PubMed), and Scopus, were reviewed for the potential role of miRNAs in the inflammatory manifestations of COVID-19. Moreover, we disclosed the interactions of inflammatory genes using STRING DB and designed interactions between miRNAs and target genes using Cityscape software. Several miRNAs, particularly miR-9, miR-98, miR-223, and miR-214, play crucial roles in the regulation of NF-kB and JAK-STAT signaling pathways as inflammatory regulators. Therefore, this group of miRNAs that mitigate inflammatory pathways can be further regarded as potential targets for far-reaching-therapeutic strategies in COVID-19 diseases.


Subject(s)
COVID-19/etiology , Inflammation/etiology , Janus Kinases/physiology , MicroRNAs/physiology , NF-kappa B/physiology , SARS-CoV-2 , STAT Transcription Factors/physiology , Humans , Signal Transduction/physiology
19.
J Zhejiang Univ Sci B ; 22(9): 782-790, 2021.
Article in English | MEDLINE | ID: mdl-34514758

ABSTRACT

Aeromonas sobria, a Gram-negative bacterium that can colonize both humans and animals, is found in a variety of environments, including water, seafood, meat, and vegetables (Cahill, 1990; Galindo et al., 2004; Song et al., 2019). Aeromonas spp. are conditionally pathogenic bacteria in aquaculture, which can rapidly proliferate, causing disease and even death in fish, especially when the environment is degraded (Neamat-Allah et al., 2020, 2021a, 2021b). In developing countries, Aeromonas spp. have been associated with a wide spectrum of infections in humans, including gastroenteritis, wound infections, septicemia, and lung infections (San Joaquin and Pickett, 1988; Wang et al., 2009; Su et al., 2013). Infections caused by Aeromonas spp. are usually more severe in immunocompromised individuals (Miyamoto et al., 2017). The presence of a plasmid encoding a ß|-lactamase in A. sobria that confers resistance to ß-lactam antibiotics poses a huge challenge to the treatment of diseases caused by this microorganism (Lim and Hong, 2020). Consequently, an in-depth understanding of the interaction between A. sobria and its hosts is urgently required to enable the development of effective strategies for the treatment of A. sobria infections.


Subject(s)
Aeromonas/pathogenicity , MAP Kinase Signaling System/physiology , Macrophages/immunology , NF-kappa B/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Cytokines/biosynthesis , Gram-Negative Bacterial Infections/immunology , Host Microbial Interactions , Mice , Signal Transduction/physiology
20.
Kaohsiung J Med Sci ; 37(12): 1113-1121, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34460994

ABSTRACT

Asthma is regarded as a chronic inflammation of the airway. Research has highlighted the significance of Vitamin D in asthma. This study explored the mechanism of vitamin D on asthma. The asthma mouse model was established by ovalbumin (OVA) sensitization and treated with vitamin D (50 or 100 ng/ml). The morphological changes of the airway were observed by HE staining. The serum IgE contents and MDA, ROS, and SOD expressions in the bronchoalveolar lavage fluid (BALF) were detected by ELISA. The Th17 and Treg cells were detected using flow cytometry. The RORγt and Foxp 3 expressions were detected by Reverse transcription quantitative polymerase chain reaction (RT-qPCR). IL-17, IL-10, and TGF-ß1 expressions were detected using ELISA. The NF-κB pathway was blocked using the NF-κB pathway inhibitor, Andrographolide sulfonate. The NF-κB pathway-related indexes were detected by western blotting. After blockade of the NF-κB pathway, the IL-17, IL-10, and TGF-G1 expressions were detected. OVA-sensitized asthma induced airway remodeling and elevated IgE content in mice, which was downregulated after vitamin D treatment. MDA and ROS were upregulated and SOD was downregulated in asthmatic mice, while vitamin D inverted the changes. Th17/Treg ratio was imbalanced, RORγt and IL-17 were upregulated, and Foxp 3, IL-10, and TGF-ß1 were downregulated after OVA sensitization, while vitamin D treatment inverted these changes and inhibited the NF-κB-p65 phosphorylation level. After blockade of the NF-κB pathway, IL-17 was downregulated and IL-10 and TGF-ß1 were upregulated. In conclusion, vitamin D rectified the Th17/Treg balance and alleviated airway inflammation by inhibiting the NF-κB pathway in asthmatic mice.


Subject(s)
Asthma/drug therapy , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Vitamin D/therapeutic use , Animals , Asthma/immunology , Asthma/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Vitamin D/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...