Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
PLoS One ; 17(2): e0261854, 2022.
Article in English | MEDLINE | ID: mdl-35108274

ABSTRACT

The nuclear factor-kappa B (NF-κB) is a transcription factor with important roles in inflammation, immune response, and oncogenesis. Dysregulation of NF-κB signaling is associated with inflammation and certain cancers. We developed a gene expression biomarker predictive of NF-κB modulation and used the biomarker to screen a large compendia of gene expression data. The biomarker consists of 108 genes responsive to tumor necrosis factor α in the absence but not the presence of IκB, an inhibitor of NF-κB. Using a set of 450 profiles from cells treated with immunomodulatory factors with known NF-κB activity, the balanced accuracy for prediction of NF-κB activation was > 90%. The biomarker was used to screen a microarray compendium consisting of 12,061 microarray comparisons from human cells exposed to 2,672 individual chemicals to identify chemicals that could cause toxic effects through NF-κB. There were 215 and 49 chemicals that were identified as putative or known NF-κB activators or suppressors, respectively. NF-κB activators were also identified using two high-throughput screening assays; 165 out of the ~3,800 chemicals (ToxCast assay) and 55 out of ~7,500 unique compounds (Tox21 assay) were identified as potential activators. A set of 32 chemicals not previously associated with NF-κB activation and which partially overlapped between the different screens were selected for validation in wild-type and NFKB1-null HeLa cells. Using RT-qPCR and targeted RNA-Seq, 31 of the 32 chemicals were confirmed to be NF-κB activators. These results comprehensively identify a set of chemicals that could cause toxic effects through NF-κB.


Subject(s)
Biomarkers/metabolism , Gene Expression Regulation/genetics , NF-kappa B/metabolism , Cell Line , Databases, Chemical , Gene Expression Regulation/drug effects , High-Throughput Screening Assays , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/deficiency , NF-kappa B p50 Subunit/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
2.
Ann Clin Lab Sci ; 51(3): 295-301, 2021 May.
Article in English | MEDLINE | ID: mdl-34162558

ABSTRACT

OBJECTIVE: Canakinumab is an interleukin (IL)-1ß inhibitory antibody. Recently, a large trial of canakinumab in cardiac patients described lower lung cancer incidence in patients treated with canakinumab compared to controls. This finding is the basis for ongoing clinical trials of canakinumab in lung cancer. To address the underlying mechanism, we established lung cancer co-cultures to investigate the interactions between lung cancer cells and immunocyte macrophages as related to the expression of IL-1ß and the effect of IL-1ß on the NF-κB pathway on lung cancer cells. METHODS: Lung cancer cell lines H838 and H1975 and macrophages were mono-cultured separately as control groups. Lung cancer cell lines and macrophages were co-cultured respectively in a ratio of 5:1 under the conditions of 37°C in a humidified atmosphere of 5% CO2 for seven days. Cell culture supernatants were collected at predetermined time points, and cell morphology was observed and photographed by microscopy. IL-1ß was detected by ELISA. H838 and H1975 cells were treated with PBS or IL-1ß for 24 hours. Cells were harvested and lysed, then analyzed in a proteome profiler array. RESULTS: Cells in co-cultures initially grew well. IL-1ß was almost undetectable in lung cancer cell lines and macrophage monoculture groups but was highly expressed in co-cultures after 24h and declined at the 7th day. In the H838 and H1975 co-culture group, the lung cancer cells occupied a great majority. IL-1ß activates the NF-κB pathway on H838 and H1975 cells. CONCLUSION: Our data showed that in a lung cancer co-culture incorporating lung cancer cells and macrophages lead to a higher expression of IL-1ß than monoculture. It is possible that such dynamic changes in IL-1ß expression may also occur in vivo in response to changes in the tumor microenvironment and interactions with immune cell populations. These interactions are likely important components to be considered when studying and modeling the expression of IL-1ß as a potential therapeutic target in lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-1beta/pharmacology , Lung Neoplasms/drug therapy , Macrophages/immunology , NF-kappa B/agonists , Tumor Microenvironment , Apoptosis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Humans , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Tumor Cells, Cultured
3.
Eur J Med Chem ; 210: 112952, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33139114

ABSTRACT

ALS is a rare type of progressive neurological disease with unknown etiology. It results in the gradual degeneration and death of motor neurons responsible for controlling the voluntary muscles. Identification of mutations in the superoxide dismutase (SOD) 1 gene has been the most significant finding in ALS research. SOD1 abnormalities have been associated with both familial as well as sporadic ALS cases. SOD2 is a highly inducible SOD that performs in concurrence with SOD1 to detoxify ROS. Induction of SOD2 can be obtained through activation of NF-Ò¡Bs. We previously reported that SRI-22819 increases NF-Ò¡B expression and activation in vitro, but it has poor ADME properties in general and has no oral bioavailability. Our initial studies were focused on direct modifications of SRI-22819. There were active compounds identified but no improvement in microsomal stability was observed. In this context, we focused on making more significant structural changes in the core of the molecule. Ataluren, an oxadiazole compound that promotes read-through and expression of dystrophin in patients with Duchenne muscular dystrophy, bears some structural similarity to SRI-22819. Thus, we synthesized a series of SRI-22819 and Ataluren (PTC124) hybrid compounds. Several compounds from this series exhibited improved activity, microsomal stability and lower calculated polar surface area (PSA). This manuscript describes the synthesis and biological evaluation of SRI-22819 analogs and its hybrid combination with Ataluren.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , NF-kappa B/agonists , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cell Line , Humans , Mice , Molecular Docking Simulation , NF-kappa B/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Structure-Activity Relationship , Superoxide Dismutase/metabolism
4.
Int J Mol Sci ; 21(22)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228049

ABSTRACT

The inhalation of metal (including lead) nanoparticles poses a real health issue to people and animals living in polluted and/or industrial areas. In this study, we exposed mice to lead(II) nitrate nanoparticles [Pb(NO3)2 NPs], which represent a highly soluble form of lead, by inhalation. We aimed to uncover the effects of their exposure on individual target organs and to reveal potential variability in the lead clearance. We examined (i) lead biodistribution in target organs using laser ablation and inductively coupled plasma mass spectrometry (LA-ICP-MS) and atomic absorption spectrometry (AAS), (ii) lead effect on histopathological changes and immune cells response in secondary target organs and (iii) the clearance ability of target organs. In the lungs and liver, Pb(NO3)2 NP inhalation induced serious structural changes and their damage was present even after a 5-week clearance period despite the lead having been almost completely eliminated from the tissues. The numbers of macrophages significantly decreased after 11-week Pb(NO3)2 NP inhalation; conversely, abundance of alpha-smooth muscle actin (α-SMA)-positive cells, which are responsible for augmented collagen production, increased in both tissues. Moreover, the expression of nuclear factor κB (NF-κB) and selected cytokines, such as tumor necrosis factor alpha (TNFα), transforming growth factor beta 1 (TGFß1), interleukin 6(IL-6), IL-1α and IL-1ß , displayed a tissue-specific response to lead exposure. In summary, diminished inflammatory response in tissues after Pb(NO3)2 NPs inhalation was associated with prolonged negative effect of lead on tissues, as demonstrated by sustained pathological changes in target organs, even after long clearance period.


Subject(s)
Air Pollutants/pharmacokinetics , Lead/pharmacokinetics , Lung/drug effects , Macrophages, Alveolar/drug effects , Metal Nanoparticles/toxicity , Nitrates/pharmacokinetics , Actins/agonists , Actins/genetics , Actins/immunology , Administration, Inhalation , Air Pollutants/toxicity , Animals , Biological Availability , Female , Gene Expression , Half-Life , Inhalation Exposure/analysis , Interleukin-1alpha/agonists , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/agonists , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/agonists , Interleukin-6/genetics , Interleukin-6/immunology , Lead/toxicity , Liver/drug effects , Liver/immunology , Liver/pathology , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Metal Nanoparticles/administration & dosage , Mice , Mice, Inbred ICR , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/immunology , Nitrates/toxicity , Spectrophotometry, Atomic , Tissue Distribution , Transforming Growth Factor beta1/agonists , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
5.
Biochem Pharmacol ; 177: 113984, 2020 07.
Article in English | MEDLINE | ID: mdl-32311348

ABSTRACT

Pluripotent stem cells are have therapeutic applications in regenerative medicine and drug discovery. However, the differentiation of stem cells in vitro hinders their large-scale production and clinical applications. The maintenance of cell pluripotency relies on a complex network of transcription factors; of these, octamer-binding transcription factor-4 (Oct4) plays a key role. This study aimed to construct an Oct4 gene promoter-driven firefly luciferase reporter and screen small-molecule compounds could maintain cell self-renewal and pluripotency. The results showed that ethyl-p-methoxycinnamate (EPMC) enhance the promoter activity of the Oct4 gene, increased the expression of Oct4 at both mRNA and protein levels, and significantly promoted the colony formation of P19 cells. These findings suggesting that EPMC could reinforce the self-renewal capacity of P19 cells. The pluripotency markers Oct4, SRY-related high-mobility-group-box protein-2, and Nanog were expressed at higher levels in EPMC-induced colonies. EPMC could promote teratoma formation and differentiation potential of P19 cells in vivo. It also enhanced self-renewal and pluripotency of human umbilical cord mesenchymal stem cells and mouse embryonic stem cells. Moreover, it significantly activated the nuclear factor kappa B (NF-κB) signaling pathway via the myeloid differentiation factor 88-dependent pathway. The expression level of Oct4 decreased after blocking the NF-κB signaling pathway, suggesting that EPMC promoted the expression of Oct4 partially through the NF-κB signaling pathway. This study indicated that EPMC could maintain self-renewal and pluripotency of stem cells.


Subject(s)
Cell Self Renewal/drug effects , Cinnamates/pharmacology , NF-kappa B/genetics , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/drug effects , Signal Transduction/drug effects , Animals , Cell Differentiation/drug effects , Cell Self Renewal/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/agonists , NF-kappa B/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/agonists , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics
6.
Development ; 147(10)2020 05 22.
Article in English | MEDLINE | ID: mdl-32291273

ABSTRACT

Retinal regeneration is robust in some cold-blooded vertebrates, but this process is ineffective in warm-blooded vertebrates. Understanding the mechanisms that suppress the reprogramming of Müller glia into neurogenic progenitors is key to harnessing the regenerative potential of the retina. Inflammation and reactive microglia are known to influence the formation of Müller glia-derived progenitor cells (MGPCs), but the mechanisms underlying this interaction are unknown. We used a chick in vivo model to investigate nuclear factor kappa B (NF-κB) signaling, a critical regulator of inflammation, during the reprogramming of Müller glia into proliferating progenitors. We find that components of the NF-κB pathway are dynamically regulated by Müller glia after neuronal damage or treatment with growth factors. Inhibition of NF-κB enhances, whereas activation suppresses, the formation of proliferating MGPCs. Following microglia ablation, the effects of NF-κB-agonists on MGPC-formation are reversed, suggesting that signals provided by reactive microglia influence how NF-κB impacts Müller glia reprogramming. We propose that NF-κB is an important signaling 'hub' that suppresses the reprogramming of Müller glia into proliferating MGPCs and this 'hub' coordinates signals provided by reactive microglia.


Subject(s)
Cell Proliferation/genetics , Chickens/growth & development , Ependymoglial Cells/metabolism , NF-kappa B/metabolism , Retina/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Animals , Cellular Reprogramming/genetics , Chickens/genetics , Gene Silencing , Intercellular Signaling Peptides and Proteins/pharmacology , Microglia/metabolism , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Neurogenesis/drug effects , Neurogenesis/genetics , Retina/growth & development , Sulfasalazine/pharmacology
7.
Eur J Pharmacol ; 873: 172981, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32014486

ABSTRACT

Dysregulated activity of the transcription factors of the nuclear factor κb (NF-κB) family has been implicated in numerous cancer types, inflammatory diseases, autoimmune disease, and other disorders. As such, selective NF-κB pathway inhibition is an attractive target to researchers for preclinical and clinical drug development. A plethora of commercially and clinically available inhibitors claim to be NF-κB specific; however, such claims of specificity are rarely quantitative or benchmarked, making the biomedical literature difficult to contextualize. This imprecision is worsened because some NF-κB reporter systems have low signal-to-noise ratios. Herein, we use a robust, defined, commercially available reporter system to benchmark NF-κB agonists and antagonists for the field. We also functionally characterize a RELA fusion-positive ependymoma cell culture with validated NF-κB inhibitor compounds.


Subject(s)
NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , Benchmarking , Cell Fusion , Cell Line, Tumor , Cell Survival , Ependymoma/pathology , HEK293 Cells , Humans , In Vitro Techniques , Reproducibility of Results , Signal-To-Noise Ratio
8.
Mar Drugs ; 17(6)2019 May 30.
Article in English | MEDLINE | ID: mdl-31151271

ABSTRACT

In our previous study, a synthetic compound, (+)-(R,E)-6a1, that incorporated the key structures of anti-inflammatory algal metabolites and the endogenous peroxisome proliferator-activated receptor γ (PPAR-γ) ligand 15-deoxy-∆12,14-prostaglandin J2 (15d-PGJ2), exerted significant PPAR-γ transcriptional activity. Because PPAR-γ expressed in macrophages has been postulated as a negative regulator of inflammation, this study was designed to investigate the anti-inflammatory effect of the PPAR-γ agonist, (+)-(R,E)-6a1. Compound (+)-(R,E)-6a1 displayed in vitro anti-inflammatory activity in lipopolysaccharides (LPS)-stimulated murine RAW264.7 macrophages. Compound (+)-(R,E)-6a1 suppressed the expression of proinflammatory factors, such as nitric oxide (NO), inducible NO synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), possibly by the inhibition of the nuclear factor-κB (NF-κB) pathway. In macrophages, (+)-(R,E)-6a1 suppressed LPS-induced phosphorylation of NF-κB, inhibitor of NF-κB α (IκBα), and IκB kinase (IKK). These results indicated that PPAR-γ agonist, (+)-(R,E)-6a1, exerts anti-inflammatory activity via inhibition of the NF-κB pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , NF-kappa B/agonists , PPAR gamma/antagonists & inhibitors , Prostaglandins, Synthetic/pharmacology , Animals , Cyclooxygenase 2/genetics , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Interleukin-6/genetics , Lipopolysaccharides , Mice , Nitric Oxide/genetics , Nitric Oxide Synthase Type II/genetics , RAW 264.7 Cells , Rhodophyta/chemistry , Tumor Necrosis Factor-alpha/genetics
9.
Biochem Pharmacol ; 166: 56-69, 2019 08.
Article in English | MEDLINE | ID: mdl-31075267

ABSTRACT

Fibromyalgia (FM) is a chronic pain syndrome involving complex interplay of biogenic amines and NMDA receptor mediated hypersensitization of nociceptive pathways. Clinical management of FM is poorly addressed with only a few available therapeutic options. Coumarins are active phenolic molecules of natural origin found to have broad pharmacological activities. Current investigation explores the role of naturally occurring coumarin, imperatorin in mouse model of fibromyalgia. Administration of reserpine (0.5 mg/kg, s.c.) thrice at 24 h intervals induced behavioral and neurochemical alterations characteristic of fibromyalgia. Reserpine was found to induce allodynia quantified using electronic von Frey (e-VF) and pressure application measurement (PAM) test, depression as indicated by an increased duration of immobility in forced swim test (FST), decreased motor coordination and locomotor activity in inclined plane test (IPT) and open field test (OFT) respectively. Cognitive deficits were evident by an increased latency to locate hidden platform in Morris water maze (MWM) and passive avoidance test (PAT). Reserpine treatment was found to cause an increased anxiety as revealed by increased time spent in closed arm of the elevated plus maze (EPM). Furthermore, an up- regulation in NMDA and NFκB expression in the brain and spinal cord was observed in reserpine treated groups. Administration of imperatorin (10 mg/kg, i.p) for a period of 5 days ameliorated all behavioral deficits, biochemical changes and decreased expression of NMDA and NFκB in the brain and spinal cord of treated mice. These findings indicate an interplay of NMDA/NFκB modulation by imperatorin in the reserpine induced fibromyalgia in mice.


Subject(s)
Fibromyalgia/drug therapy , Fibromyalgia/metabolism , Furocoumarins/therapeutic use , N-Methylaspartate/metabolism , NF-kappa B/metabolism , Reserpine/toxicity , Adrenergic Uptake Inhibitors/toxicity , Animals , Dose-Response Relationship, Drug , Fibromyalgia/chemically induced , Furocoumarins/pharmacology , Mice , N-Methylaspartate/agonists , N-Methylaspartate/antagonists & inhibitors , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Treatment Outcome
10.
Food Funct ; 9(9): 4865-4875, 2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30160278

ABSTRACT

Osteoarthritis (OA) is characterized by the degeneration and destruction of articular cartilage. Allicin, a dietary garlic active constituent, exerts anti-inflammatory effects on several diseases. However, its effects on OA have not been clearly elucidated. In this study, we explored the effects of allicin on OA in both in vitro and in vivo models. Allicin inhibited interleukin-1ß (IL-1ß) induced overproduction of nitric oxide, inducible nitric oxide synthase, prostaglandin E2, and cyclooxygenase-2, as well as pro-inflammatory cytokines tumor necrosis factor alpha and interleukin-6 in chondrocytes in a dose-dependent manner. Meanwhile, allicin reversed the overproduction of metalloproteinase-13 and a disintegrin and metalloproteinase with thrombospondin motifs-5 and the decrease of aggrecan and type II collagen. Furthermore, allicin dramatically suppressed IL-1ß-stimulated PI3K/Akt/NF-κB activation in chondrocytes. In vivo, treatment with allicin prevented the destruction of cartilage and inhibited PI3K/Akt/NF-κB activation in the cartilage of mice OA models. Taken together, these results indicate that allicin may be a potential therapeutic agent for OA.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Chondrocytes/metabolism , Dietary Supplements , Disease Models, Animal , Osteoarthritis, Knee/therapy , Phosphoinositide-3 Kinase Inhibitors , Sulfinic Acids/therapeutic use , Animals , Bone Density Conservation Agents/adverse effects , Bone Density Conservation Agents/metabolism , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/pathology , Dietary Supplements/adverse effects , Disease Progression , Disulfides , Female , Gene Expression Regulation , Humans , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/physiopathology , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Sulfinic Acids/adverse effects , Sulfinic Acids/metabolism
11.
Toxicol Lett ; 294: 44-50, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29753068

ABSTRACT

As widely used in consumer products, perfluorooctanoic acid (PFOA) has become a common environmental pollutant, which has been detected in human serum and associated with cancers. Our previous study showed that PFOA is a carcinogen that promotes endometrial cancer cell migration and invasion through activation of ERK/mTOR signaling. Here, we showed that PFOA (≥100 nM) treatment also stimulated A2780 ovarian cancer cell invasion and migration, which correlated with increased matrix metalloproteinases MMP-2/-9 expression, important proteases associated with tumor invasion and migration. Notably, PFOA treatment induced activation of ERK1/2/ NF-κB signaling. Pre-treatment with U0126, an ERK1/2inhibitor;or JSH-23, a NF-kB inhibitor, can reverse the PFOA-induced cell migration and invasion. Consistent with these results, inhibiting ERK1/2 or NF-κB signaling abolished PFOA-induced up-regulation of MMP-2/-9 expression. These results indicate that PFOA can stimulate ovarian cancer cell migration, invasion and MMP-2/-9 expression by up-regulating ERK/NF-κB pathway.


Subject(s)
Caprylates/toxicity , Carcinogens, Environmental/toxicity , Fluorocarbons/toxicity , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/agonists , Ovarian Neoplasms/chemically induced , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Butadienes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Induction/drug effects , Female , Humans , Kinetics , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nitriles/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phenylenediamines/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects
12.
Biomed Pharmacother ; 103: 490-498, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29677534

ABSTRACT

Papillary thyroid carcinoma (PTC) is the most common endocrine carcinoma. Our previous study revealed that punicalagin (PUN), an active component from pomegranate, triggered autophagic cell death and DNA damage response (DDR) in papillary thyroid carcinoma BCPAP cells. But the detailed anti-cancer mechanisms of punicalagin against PTC still remained to be further explored. DDR activation is a proven cause of cellular senescence, which mediates anti-tumor processes under certain circumstances. In this study, we reported that punicalagin treatment generated a senescent phenotype of BCPAP cells characterized as altered morphology, increased cell granularity and senescence-associated ß-galactosidase (SA-ß-Gal) staining. Senescence induced by punicalagin treatment was further confirmed by cell cycle arrest and upregulation of cyclin-dependent kinase inhibitor p21. Meanwhile, the senescence-associated secretory phenotype (SASP) included high levels of inflammatory cytokines, principally IL-6 and IL-1ß. Furthermore, punicalagin exposure caused the phosphorylation and subsequent degradation of IκBα as well as the nuclear translocation of p65, suggesting the activation of NF-κB signaling pathway. Inhibition of NF-κB by pyrrolidine dithiocarbamate (PDTC), a selective inhibitor of NF-κB, partially reversed the cellular senescent phenotype induced by punicalagin in BCPAP cells as evidenced by the decreased fraction of SA-ß-Gal staining positive cells and blockage of SASP generation. These results collectively showed that punicalagin treatment induced senescent growth arrest and SASP via triggering NF-κB activation. These observations elucidated novel anti-cancer mechanisms of punicalagin and might provide new potential prospects for PTC therapy.


Subject(s)
Carcinoma, Papillary/metabolism , Cell Cycle Checkpoints/drug effects , Cellular Senescence/drug effects , Hydrolyzable Tannins/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Thyroid Neoplasms/metabolism , Carcinoma, Papillary/drug therapy , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cellular Senescence/physiology , Dose-Response Relationship, Drug , Humans , Hydrolyzable Tannins/therapeutic use , NF-kappa B/agonists , Signal Transduction/physiology , Thyroid Cancer, Papillary , Thyroid Neoplasms/drug therapy
13.
Arch Physiol Biochem ; 124(5): 468-474, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29364751

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the effects of high glucose (HG) on inflammation in HepG2 cells. METHODS: The molecular mechanisms linking HG to inflammation was assessed in HepG2 cells exposed to HG (33 mM). RESULTS: The results showed that HG significantly enhanced TNF-α, IL-6 and PAI-1 expression in HepG2 cells. Increased expression of cytokines was accompanied by enhanced phosphorylation of JNK, P38, ERK and IKKα/IKKß. In addition, JNK, ERK, P38 and NF-kB inhibitors could significantly attenuate HG-induced expression of TNF-α, IL-6 and PAI-1. Furthermore, HG could promote the generation of reactive oxygen species (ROS), while N-acetyl cysteine, a ROS scavenger, had an inhibitory effect on the expression of TNF-α, IL-6 and PAI-1 in HG-treated cells. CONCLUSIONS: Our results indicated that HG-induced inflammation is mediated through the generation of ROS and activation of the MAPKs and NF-kB signalling pathways in HepG2 cells.


Subject(s)
Gene Expression Regulation , Hepatocytes/metabolism , Hyperglycemia/metabolism , MAP Kinase Signaling System , NF-kappa B/agonists , Oxidative Stress , Tumor Necrosis Factor-alpha/agonists , Acetylcysteine/pharmacology , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Glucose/adverse effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Hyperglycemia/immunology , Hyperglycemia/pathology , Interleukin-6/agonists , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Osmolar Concentration , Oxidative Stress/drug effects , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/agonists , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Reproducibility of Results , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
J Biol Chem ; 293(8): 2701-2710, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29301940

ABSTRACT

Mammalian Nod-like receptor (NLR) proteins contribute to the regulation and induction of innate and adaptive immunity in mammals, although the function of about half of the currently identified NLR proteins remains poorly characterized. Here we analyzed the function of the primate-specific NLRP11 gene product. We show that NLRP11 is highly expressed in immune cells, including myeloid cells, B cells, and some B cell lymphoma lines. Overexpression of NLRP11 in human cells did not trigger key innate immune signaling pathways, including NF-κB and type I interferon responses. NLRP11 harbors a pyrin domain, which is responsible for inflammasome formation in related NLR proteins. However, NLRP11 did not interact with the inflammasome adaptor protein ASC, and it did not trigger caspase-1 activation. By contrast, expression of NLRP11 specifically repressed NF-κB and type I interferon responses, two key innate immune pathways involved in inflammation. This effect was independent of the pyrin domain and ATPase activity of NLRP11. siRNA-mediated knockdown of NLRP11 in human myeloid THP1 cells validated these findings and revealed enhanced lipopolysaccharide and Sendai virus-induced cytokine and interferon responses, respectively, in cells with reduced NLRP11 expression. In summary, our work identifies a novel role of NLRP11 in the regulation of inflammatory responses in human cells.


Subject(s)
B-Lymphocytes/metabolism , Down-Regulation , Gene Expression Regulation , Immunity, Innate , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid Cells/metabolism , NLR Proteins/metabolism , Amino Acid Substitution , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Line, Transformed , Cell Line, Tumor , Down-Regulation/drug effects , Female , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Humans , Immunity, Innate/drug effects , Interferon Type I/agonists , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/toxicity , Male , Mutation , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/immunology , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , NLR Proteins/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Organ Specificity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
15.
Mol Med Rep ; 17(2): 2501-2508, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207194

ABSTRACT

Highly active antiretroviral therapy (HAART) is very effective in suppressing human immunodeficiency virus type 1 (HIV­1) replication. However, the treatment is required to be administered for the remainder of an individual's lifetime due to latent HIV­1 reservoirs. The 'shock­and­kill' strategy, which involves using agents to reactivate latent HIV­1 and subsequently killing latently infected cells in the presence of HAART, was recently proposed. Unfortunately, no agents have currently demonstrated an ability to reactivate latent HIV­1 in vivo in the absence of toxicity. Therefore, the identification of novel latency activators is required. In order to identify a potential novel agent, the present study investigated the effect of quercetin on latent HIV­1 reactivation using an established model of HIV­1 latency. As a marker for reactivation of HIV­1 in C11 Jurkat cells, the expression of green fluorescent protein, controlled by HIV­1 long terminal repeat, was observed by fluorescence microscopy. The results of the present study demonstrated that quercetin effectively reactivated latent HIV­1 gene expression alone, and led to synergistic reactivation when combined with prostratin or valproic acid. In addition, the present study provides evidence that quercetin may reactivate HIV­1 expression by inducing nuclear factor­κB nuclear translocation, and that the toxicity of quercetin is lower when compared with various additional activators of HIV­1. Combined, the results of the present study indicate that quercetin may be an effective agent to disrupt HIV­1 latency and may be useful in future eradication strategies.


Subject(s)
HIV Infections/metabolism , HIV Infections/virology , HIV-1/drug effects , HIV-1/physiology , NF-kappa B/agonists , Quercetin/pharmacology , Virus Latency/drug effects , Cell Line , Humans , Jurkat Cells , Virus Activation/drug effects , Virus Activation/genetics , Virus Replication/drug effects
16.
J Biochem Mol Toxicol ; 32(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-29286200

ABSTRACT

Proximal tubule protein take-up is interceded by 2 receptors, megalin and cubilin. These receptors rescue an assortment of filtered ligands including fundamental vitamins and hormones. The objective of this study was to investigate the potential relation of megalin receptor injury with nuclear factor-kappa B (NF-κB) upregulation in acute kidney injury rat model. Twenty four rats were allocated into two groups: control group received saline, while the second group was intoxicated with cadmium chloride (2.4 mg Cd/kg/day i.p) for 30 days. Blood urea nitrogen, serum creatinine, tissue oxidant-antioxidant parameters (malondialdehyde [MDA] and reduced glutathione [GSH]) and expression levels for NF-κB, toll like receptor-2 (TLR2), toll like receptor-4 (TLR4), and megalin receptor were estimated. Noticeable downregulation of megalin receptor versus upregulation of NF-κB, TLR2, and TLR4 were observed in AKI rat model together with significant elevation in MDA as well as significant reduction in GSH. The study concluded that the oxidative stress in kidney tissue leads to megalin receptor damage, which indeed motivates upregulation of NF-κB through TLRs 2 and 4 pathways.


Subject(s)
Acute Kidney Injury/etiology , Cadmium Poisoning/physiopathology , Gene Expression Regulation/drug effects , Kidney/drug effects , Low Density Lipoprotein Receptor-Related Protein-2/antagonists & inhibitors , NF-kappa B/agonists , Oxidative Stress/drug effects , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Animals , Biomarkers/blood , Biomarkers/metabolism , Cadmium Chloride/toxicity , Glutathione/antagonists & inhibitors , Glutathione/chemistry , Glutathione/metabolism , Kidney/metabolism , Kidney/physiopathology , Lipid Peroxidation/drug effects , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Malondialdehyde/agonists , Malondialdehyde/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidation-Reduction , Random Allocation , Rats, Sprague-Dawley , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
17.
J Biol Chem ; 293(8): 2661-2674, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29196608

ABSTRACT

TRIM5α is an important host restriction factor that could potently block retrovirus infection. The SPRY domain of TRIM5α mediates post-entry restriction by recognition of and binding to the retroviral capsid. Human TRIM5α also functions as an innate immune sensor to activate AP-1 and NF-κB signaling, which subsequently restrict virus replication. Previous studies have shown that the AP-1 and NF-κB signaling activation relies on the RING motif of TRIM5α. In this study, we have demonstrated that the SPRY domain is essential for rhesus macaque TRIM5α to activate AP-1 but not NF-κB signaling. The AP-1 activation mainly depends on all of the ß-sheet barrel on SPRY structure of TRIM5α. Furthermore, the SPRY-mediated auto-ubiquitination of TRIM5α is required for AP-1 activation. This study reports that rhesus macaque TRIM5α mainly undergoes Lys27-linked and Met1-linked auto-polyubiquitination. Finally, we found that the TRIM5α signaling function was positively correlated with its retroviral restriction activity. This study discovered an important role of the SPRY domain in immune signaling and antiviral activity and further expanded our knowledge of the antiviral mechanism of TRIM5α.


Subject(s)
B30.2-SPRY Domain , Models, Molecular , Replication Protein C/metabolism , Signal Transduction , Tripartite Motif Proteins/metabolism , Ubiquitination , Animals , Enzyme Activation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Macaca fascicularis , Macaca mulatta , NF-kappa B/agonists , NF-kappa B/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation, beta-Strand , RING Finger Domains , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Replication Protein C/chemistry , Replication Protein C/genetics , Species Specificity , Tripartite Motif Proteins/chemistry , Tripartite Motif Proteins/genetics
18.
Asian J Androl ; 20(1): 24-29, 2018.
Article in English | MEDLINE | ID: mdl-28382926

ABSTRACT

Emerging evidence indicates that aldosterone and mineralocorticoid receptors (MRs) are associated with the pathogenesis of erectile dysfunction. However, the molecular mechanisms remain largely unknown. In this study, freshly isolated penile corpus cavernosum tissue from rats was treated with aldosterone, with or without MRs inhibitors. Nuclear factor (NF)-kappa B (NF-κB) activity was evaluated by real-time quantitative PCR, luciferase assay, and immunoblot. The results demonstrated that mRNA levels of the NF-κB target genes, including inhibitor of NF-κB alpha (IκB-α), NF-κB1, tumor necrosis factor-alpha (TNF-α), and interleukin 6 (IL-6), were higher after aldosterone treatment. Accordingly, phosphorylation of p65/RelA, IκB-α, and inhibitor of NF-κB kinase-ß was markedly increased by aldosterone. Furthermore, knockdown of MRs prevented activation of the NF-κB canonical pathway by aldosterone. Consistent with this finding, ectopic overexpression of MRs enhanced the transcriptional activation of NF-κB by aldosterone. More importantly, the MRs antagonist, spironolactone blocked aldosterone-mediated activation of the canonical NF-κB pathway. In conclusion, aldosterone has an inflammatory effect in the corpus cavernosum penis, inducing NF-κB activation via an MRs-dependent pathway, which may be prevented by selective MRs antagonists. These data reveal the possible role of aldosterone in erectile dysfunction as well as its potential as a novel pharmacologic target for treatment.


Subject(s)
Aldosterone/pharmacology , Cytokines/biosynthesis , NF-kappa B/agonists , Penis/metabolism , Signal Transduction/drug effects , Animals , Gene Knockdown Techniques , I-kappa B Kinase/antagonists & inhibitors , Interleukin-6/biosynthesis , Interleukin-6/genetics , Male , Mineralocorticoid Receptor Antagonists/pharmacology , NF-kappa B/genetics , Penis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rats , Rats, Inbred WKY , Receptors, Mineralocorticoid/biosynthesis , Receptors, Mineralocorticoid/genetics , Spironolactone/pharmacology , Transcriptional Activation , Tumor Necrosis Factor-alpha/biosynthesis , NF-kappaB-Inducing Kinase
19.
Cell Death Dis ; 8(8): e3024, 2017 08 31.
Article in English | MEDLINE | ID: mdl-29048425

ABSTRACT

Cardamonin has been demonstrated to have an inhibitory effect in many cancers, but its underlying mechanism remains elusive. Here, we studied, for the first time, the mechanism of cardamonin-induced nasopharyngeal carcinoma cell death both in vitro and in vivo. In our study, we showed that cardamonin inhibited cancer cell growth by inducing G2/M phase cell cycle arrest and apoptosis via accumulation of ROS. NF-κB activation was involved in breaking cellular redox homeostasis. Therefore, our results provided new insight into the mechanism of the antitumor effect of cardamonin, supporting cardamonin as a prospective therapeutic drug in nasopharyngeal carcinoma by modulating intracellular redox balance.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/drug therapy , Chalcones/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , NF-kappa B/genetics , Nasopharyngeal Neoplasms/drug therapy , Animals , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Mice, Nude , NF-kappa B/agonists , NF-kappa B/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
Biochem Soc Trans ; 45(4): 953-62, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28687714

ABSTRACT

Termed 'master gene regulators' long ncRNAs (lncRNAs) have emerged as the true vanguard of the 'noncoding revolution'. Functioning at a molecular level, in most if not all cellular processes, lncRNAs exert their effects systemically. Thus, it is not surprising that lncRNAs have emerged as important players in human pathophysiology. As our body's first line of defense upon infection or injury, inflammation has been implicated in the etiology of several human diseases. At the center of the acute inflammatory response, as well as several pathologies, is the pleiotropic transcription factor NF-κß. In this review, we attempt to capture a summary of lncRNAs directly involved in regulating innate immunity at various arms of the NF-κß pathway that have also been validated in human disease. We also highlight the fundamental concepts required as lncRNAs enter a new era of diagnostic and therapeutic significance.


Subject(s)
Chronic Disease , Gene Expression Regulation, Developmental , Immunity, Innate , Models, Immunological , RNA, Long Noncoding/metabolism , Signal Transduction , Animals , Genetic Predisposition to Disease , Humans , Inflammation Mediators/agonists , Inflammation Mediators/metabolism , NF-kappa B/agonists , NF-kappa B/genetics , NF-kappa B/metabolism , Polymorphism, Single Nucleotide , RNA, Long Noncoding/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...