Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38338683

ABSTRACT

MicroRNAs (miRNAs) are involved in the modulation of pathogenic genes by binding to their mRNA sequences' 3' untranslated regions (3'UTR). Interleukin-6 (IL-6) is known to promote cancer progression and treatment resistance. In this study, we aimed to explore the therapeutic effects of gold nanoparticles (GNP) against IL-6 overexpression and the modulation of miRNA-26a-5p in breast cancer (BC) cells. GNP were synthesized using the trisodium citrate method and characterized through UV-Vis spectroscopy, dynamic light scattering (DLS), and transmission electron microscopy (TEM). To predict the binding of miR-26a-5p in the IL-6 mRNA's 3'UTR, we utilized bioinformatics algorithms. Luciferase reporter clone assays and anti-miRNA-26a-5p transfection were employed to validate the binding of miR26a-5p in the IL-6 mRNA's 3'UTR. The activity of RelA and NF-κBp50 was assessed and confirmed using Bay 11-7082. The synthesized GNP were spherical with a mean size of 28.3 nm, exhibiting high stability, and were suitable for BC cell treatment. We found that miR-26a-5p directly regulated IL-6 overexpression in MCF-7 cells activated with PMA. Treatment of MCF-7 cells with GNP resulted in the inhibition of IL-6 overexpression and secretion through the increase of miR26a-5p. Furthermore, GNP deactivated NF-κBp65/NF-κBp50 transcription activity. The newly engineered GNP demonstrated safety and showed promise as a therapeutic approach for reducing IL-6 overexpression. The GNP suppressed IL-6 overexpression and secretion by deactivating NF-κBp65/NF-κBp50 transcription activity and upregulating miR-26a-5p expression in activated BC cells. These findings suggest that GNP have potential as a therapeutic intervention for BC by targeting IL-6 expression and associated pathways.


Subject(s)
Breast Neoplasms , Metal Nanoparticles , MicroRNAs , NF-kappa B , Female , Humans , 3' Untranslated Regions , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gold , Interleukin-6/genetics , Interleukin-6/metabolism , Metal Nanoparticles/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism
2.
J Neurochem ; 157(3): 574-585, 2021 05.
Article in English | MEDLINE | ID: mdl-33289070

ABSTRACT

Nuclear factor (NF)-κB-mediated neuroinflammation is an important mechanism of intracerebral hemorrhage (ICH)-induced neurotoxicity. Silent information regulator 1 (SIRT1) plays a multi-protective effect in a variety of diseases by deacetylating and inhibiting NF-κB/p65. However, the role of SIRT1 in brain damage following ICH remains unclear. We hypothesized that SIRT1 can protect against ICH-induced brain damage by inhibiting neuroinflammation through deacetylating NF-κB/p65. The ICH model was induced in vivo (with collagenase) and in vitro (with hemoglobin). Resveratrol and Ex527 were administered to activate or inhibit SIRT1, respectively. Western blot, immunohistochemistry, and immunofluorescence assays were performed to detect the expression of SIRT1 and p65. Enzyme-linked immunosorbent assays (ELISAs) were used to explore tumor necrosis factor (TNF)-α and interleukin (IL)-1ß release. The neurological score, brain water content, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and brain hemoglobin content were determined to evaluate the neuroprotective effect of SIRT1. SIRT1 expression was decreased, whereas the level of acetylated p65 (Ac-p65) was elevated after ICH in vivo. Moreover, hemoglobin treatment decreased the expression of SIRT1 in vitro. Activation of SIRT1 by resveratrol had a neuroprotective effect, along with decreased levels of Ac-p65, IL-1ß, TNF-α, and apoptosis after ICH. The effect of resveratrol was abolished by the SIRT1 inhibitor Ex527. Our results are consistent with the hypothesis that SIRT1 exerts a neuroprotective effect after ICH by deacetylating p65 to inhibit the NF-κB-dependent inflammatory response.


Subject(s)
Cerebral Hemorrhage/drug therapy , Neuroprotective Agents , Sirtuin 1/genetics , Transcription Factor RelA/drug effects , Acetylation , Animals , Apoptosis/drug effects , Cerebral Hemorrhage/chemically induced , Collagenases , Encephalitis/drug therapy , Encephalitis/pathology , Hemoglobins , Injections, Intraventricular , Interleukin-1beta/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Primary Cell Culture , Resveratrol/therapeutic use , Sirtuin 1/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Neurobiol Learn Mem ; 182: 107463, 2021 07.
Article in English | MEDLINE | ID: mdl-34015440

ABSTRACT

Growing evidences indicate that neuropathic pain is frequently accompanied with cognitive impairments, which aggravate the decrease in the quality of life of chronic pain patients. Furthermore, it has been shown that the activation of Glucagon-like-peptide-1receptor (GLP-1R) improved memory deficit in multiple diseases, including Alzheimer's disease (AD), stroke. However, whether GLP-1R activation could improve memory impairment induced by neuropathic pain and the mechanisms underlying the effect of the activation of GLP-1R on memory protection have not yet been established. The spared nerve injury (SNI) model was established as a kind of neuropathic pain. And novel-object recognition memory (hippocampus-dependent memory) was tested by the novel object recognition test (NORT). The expression levels of GLP-1, GLP-1R, adenosine monophosphate-activated protein kinase (AMPK), p-AMPKThr172, nuclear factor κ B p65 (NF-κB p65), interleukin-1beta (IL-1ß), IL-1ß p17 (mature IL-1ß), tumor necrosis factor-alpha (TNF-α) and the synaptic proteins were tested in the murine hippocampus with memory deficits caused by neuropathic pain. Then, exenatide acetate (Ex-4, a GLP-1R agonist), exendin (9-39) (Ex(9-39), a GLP-1R antagonist) and Compound C dihydrochloride (CC, an AMPK inhibitor) were used to test the effects of the activation of GLP-1R in the mice with neuropathic pain. First, we uncovered that neuropathic pain could inhibit GLP-1/GLP-R axis, disturb inflammatory signaling pathway, increase the expression of IL-1ß, IL-1ß p17 and TNF-α, downregulate the synaptic proteins (postsynaptic density protein 95 (PSD95) and Arc). Subsequently, we reported that Ex-4 treatment could improve recognition memory impairment, increase the ratio of p-AMPKThr172/AMPK, inhibit the phosphorylation NF-κB p65 and decrease the expression of IL-1ß, IL-1ß p17 and TNF-α, upregulate the levels of PSD95 and Arc. Moreover, we found that Ex(9-39) and CC treatment could abrogate the memory protection of activation of GLP-1R in mice with neuropathic pain. The results indicated that the activation of GLP-1R could improve recognition memory impairment via regulating AMPK/NF-κB pathway, improving neuroinflammation, reversing the decreased level of synaptic proteins in neuropathic pain mice.


Subject(s)
AMP-Activated Protein Kinase Kinases/drug effects , Exenatide/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Hippocampus/drug effects , Neuralgia/metabolism , Recognition, Psychology/drug effects , Transcription Factor RelA/drug effects , AMP-Activated Protein Kinase Kinases/metabolism , Animals , Chronic Pain/metabolism , Chronic Pain/physiopathology , Disease Models, Animal , Glucagon-Like Peptide 1/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Hippocampus/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Neuralgia/physiopathology , Neuroinflammatory Diseases/metabolism , Open Field Test , Peptide Fragments/pharmacology , Peripheral Nerve Injuries , Sciatic Nerve/surgery , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
4.
Am J Respir Crit Care Med ; 201(8): 934-945, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31834999

ABSTRACT

Rationale: The role of FSTL-1 (follistatin-like 1) in lung homeostasis is unknown.Objectives: We aimed to define the impact of FSTL-1 attenuation on lung structure and function and to identify FSTL-1-regulated transcriptional pathways in the lung. Further, we aimed to analyze the association of FSTL-1 SNPs with lung disease.Methods: FSTL-1 hypomorphic (FSTL-1 Hypo) mice underwent lung morphometry, pulmonary function testing, and micro-computed tomography. Fstl1 expression was determined in wild-type lung cell populations from three independent research groups. RNA sequencing of wild-type and FSTL-1 Hypo mice identified FSTL-1-regulated gene expression, followed by validation and mechanistic in vitro examination. FSTL1 SNP analysis was performed in the COPDGene (Genetic Epidemiology of Chronic Obstructive Pulmonary Disease) cohort.Measurements and Main Results: FSTL-1 Hypo mice developed spontaneous emphysema, independent of smoke exposure. Fstl1 is highly expressed in the lung by mesenchymal and endothelial cells but not immune cells. RNA sequencing of whole lung identified 33 FSTL-1-regulated genes, including Nr4a1, an orphan nuclear hormone receptor that negatively regulates NF-κB (nuclear factor-κB) signaling. In vitro, recombinant FSTL-1 treatment of macrophages attenuated NF-κB p65 phosphorylation in an Nr4a1-dependent manner. Within the COPDGene cohort, several SNPs in the FSTL1 region corresponded to chronic obstructive pulmonary disease and lung function.Conclusions: This work identifies a novel role for FSTL-1 protecting against emphysema development independent of smoke exposure. This FSTL-1-deficient emphysema implicates regulation of immune tolerance in lung macrophages through Nr4a1. Further study of the mechanisms involving FSTL-1 in lung homeostasis, immune regulation, and NF-κB signaling may provide additional insight into the pathophysiology of emphysema and inflammatory lung diseases.


Subject(s)
Follistatin-Related Proteins/genetics , Lung/diagnostic imaging , Pulmonary Emphysema/genetics , Smoke/adverse effects , Animals , Endothelial Cells/metabolism , Follistatin-Related Proteins/pharmacology , Gene Expression Regulation , Gene Knockdown Techniques , Humans , In Vitro Techniques , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphorylation/drug effects , Polymorphism, Single Nucleotide , Positron Emission Tomography Computed Tomography , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Emphysema/diagnostic imaging , Pulmonary Emphysema/metabolism , Single Photon Emission Computed Tomography Computed Tomography , Nicotiana , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , X-Ray Microtomography
5.
Rheumatology (Oxford) ; 59(4): 742-753, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31377809

ABSTRACT

OBJECTIVES: Xerostomia in SS patients has been associated with low quality and quantity of salivary mucins, which are fundamental for the hydration and protection of the oral mucosa. The aim of this study was to evaluate if cytokines induce aberrant mucin expression and whether tauroursodeoxycholic acid (TUDCA) is able to counteract such an anomaly. METHODS: Labial salivary glands from 16 SS patients and 15 control subjects, as well as 3D acini or human submandibular gland cells stimulated with TNF-α or IFN-γ and co-incubated with TUDCA, were analysed. mRNA and protein levels of Mucin 1 (MUC1) and MUC7 were determined by RT-qPCR and western blot, respectively. Co-immunoprecipitation and immunofluorescence assays for mucins and GRP78 [an endoplasmic reticulum (ER)-resident protein] were also performed. mRNA levels of RelA/p65 (nuclear factor-κB subunit), TNF-α, IL-1ß, IL-6, SEL1L and EDEM1 were determined by RT-qPCR, and RelA/p65 localization was evaluated by immunofluorescence. RESULTS: MUC1 is overexpressed and accumulated in the ER of labial salivary gland from SS patients, while MUC7 accumulates throughout the cytoplasm of acinar cells; however, MUC1, but not MUC7, co-precipitated with GRP78. TUDCA diminished the overexpression and aberrant accumulation of MUC1 induced by TNF-α and IFN-γ, as well as the nuclear translocation of RelA/p65, together with the expression of inflammatory and ER stress markers in 3D acini. CONCLUSION: Chronic inflammation alters the secretory process of MUC1, inducing ER stress and affecting the quality of saliva in SS patients. TUDCA showed anti-inflammatory properties decreasing aberrant MUC1 accumulation. Further studies are necessary to evaluate the potential therapeutic effect of TUDCA in restoring glandular homeostasis in SS patients.


Subject(s)
Acinar Cells/drug effects , Endoplasmic Reticulum Stress/drug effects , Mucin-1/drug effects , Salivary Glands, Minor/drug effects , Sjogren's Syndrome/metabolism , Submandibular Gland/drug effects , Taurochenodeoxycholic Acid/pharmacology , Xerostomia/metabolism , Acinar Cells/metabolism , Adult , Aged , Case-Control Studies , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Female , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Immunoprecipitation , In Vitro Techniques , Interferon-gamma/pharmacology , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Mucin-1/genetics , Mucin-1/metabolism , Mucins/drug effects , Mucins/genetics , Mucins/metabolism , Proteins/drug effects , Proteins/genetics , Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Salivary Glands, Minor/metabolism , Salivary Proteins and Peptides/drug effects , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/metabolism , Sjogren's Syndrome/genetics , Submandibular Gland/cytology , Submandibular Gland/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Xerostomia/genetics
6.
Toxicol Appl Pharmacol ; 391: 114915, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32035082

ABSTRACT

Idiosyncratic drug-induced liver injury (IDILI) is a severe disease that cannot be detected during drug development. It has been shown that hepatotoxicity of some compounds associated with IDILI becomes apparent when these are combined in vivo and in vitro with LPS or TNF. Among these compounds trovafloxacin (TVX) induced apoptosis in the liver and increased pro-inflammatory cytokines in mice exposed to LPS/TNF. The hepatocyte survival and the cytokine release after TNF/LPS stimulation relies on a pulsatile activation of NF-κB. We set out to evaluate the dynamic activation of NF-κB in response to TVX + TNF or LPS models, both in mouse and human cells. Remarkably, TVX prolonged the first translocation of NF-κB induced by TNF both in vivo and in vitro. The prolonged p65 translocation caused by TVX was associated with an increased phosphorylation of IKK and MAPKs and accumulation of inhibitors of NF-κB such as IκBα and A20 in HepG2. Coherently, TVX suppressed further TNF-induced NF-κB translocations in HepG2 leading to decreased transcription of ICAM-1 and inhibitors of apoptosis. TVX prolonged LPS-induced NF-κB translocation in RAW264.7 macrophages increasing the secretion of TNF. In summary, this study presents new, relevant insights into the mechanism of TVX-induced liver injury underlining the resemblance between mouse and human models. In this study we convincingly show that regularly used toxicity models provide a coherent view of relevant pathways for IDILI. We propose that assessment of the kinetics of activation of NF-κB and MAPKs is an appropriate tool for the identification of hepatotoxic compounds during drug development.


Subject(s)
Anti-Bacterial Agents/toxicity , Chemical and Drug Induced Liver Injury/pathology , Fluoroquinolones/toxicity , Lipopolysaccharides/pharmacology , Naphthyridines/toxicity , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Translocation, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/genetics , Cytokines/metabolism , Humans , I-kappa B Proteins/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
7.
FASEB J ; 32(9): 5051-5062, 2018 09.
Article in English | MEDLINE | ID: mdl-29641288

ABSTRACT

Angiogenic factor with G-patch and FHA domains 1 (AGGF1) is involved in vascular development, angiogenesis, specification of hemangioblasts, and differentiation of veins. When mutated, however, it causes Klippel-Trenaunay syndrome, a vascular disorder. In this study, we show that angiotensin II (AngII)-the major effector of the renin-angiotensin system and one of the most important regulators of the cardiovascular system-induces the expression of AGGF1 through NF-κB, and that AGGF1 plays a key role in AngII-induced angiogenesis. AngII significantly up-regulated the levels of AGGF1 mRNA and protein in HUVECs at concentrations of 10-40 µg/ml but not >60 µg/ml. AngII type 1 receptor (AT1R) inhibitor losartan inhibited AngII-induced up-regulation of AGGF1, whereas AT2R inhibitor PD123319 further increased AngII-induced up-regulation of AGGF1. Up-regulation of AGGF1 by AngII was blocked by NF-κB inhibitors, and p65 binds directly to a binding site at the promoter/regulatory region of AGGF1 and transcriptionally activates AGGF1 expression. AngII-induced endothelial tube formation was blocked by small interfering RNAs (siRNAs) for RELA (RELA proto-oncogene, NF-κB subunit)/p65 or AGGF1, and the effect of RELA siRNA was rescued by AGGF1. AngII-induced angiogenesis from aortic rings was severely impaired in Aggf1+/- mice, and the effect was restored by AGGF1. These data suggest that AngII acts as a critical regulator of AGGF1 expression through NF-κB, and that AGGF1 plays a key role in AngII-induced angiogenesis.-Si, W., Xie, W., Deng, W., Xiao, Y., Karnik, S. S., Xu, C., Chen, Q., Wang, Q. K. Angiotensin II increases angiogenesis by NF-κB-mediated transcriptional activation of angiogenic factor AGGF1.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Angiogenic Proteins/metabolism , Angiotensin II/pharmacology , NF-kappa B/drug effects , Transcriptional Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Imidazoles/pharmacology , Losartan/pharmacology , NF-kappa B/metabolism , Neovascularization, Pathologic/drug therapy , Proto-Oncogene Mas , Pyridines/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , Transcription Factor RelA/drug effects
8.
Molecules ; 24(4)2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30781671

ABSTRACT

Cancer is still a major risk factor to public health globally, causing approximately 9.8 million deaths worldwide in 2018. Despite advances in conventional treatment modalities for cancer treatment, there are still few effective therapies available due to the lack of selectivity, adverse side effects, non-specific toxicities, and tumour recurrence. Therefore, there is an immediate need for essential alternative therapeutics, which can prove to be beneficial and safe against cancer. Various phytochemicals from natural sources have been found to exhibit beneficial medicinal properties against various human diseases. Zerumbone is one such compound isolated from Zingiber zerumbet Smith that possesses diverse pharmacological properties including those of antioxidant, antibacterial, antipyretic, anti-inflammatory, immunomodulatory, as well as anti-neoplastic. Zerumbone has shown its anti-cancer effects by causing significant suppression of proliferation, survival, angiogenesis, invasion, and metastasis through the molecular modulation of different pathways such as NF-κB, Akt, and IL-6/JAK2/STAT3 (interleukin-6/janus kinase-2/signal transducer and activator of transcription 3) and their downstream target proteins. The current review briefly summarizes the modes of action and therapeutic potential of zerumbone against various cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Sesquiterpenes/administration & dosage , Zingiberaceae/chemistry , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Interleukin-6/metabolism , Janus Kinase 2/metabolism , Molecular Structure , STAT3 Transcription Factor/drug effects , Sesquiterpenes/adverse effects , Signal Transduction , Transcription Factor RelA/drug effects
9.
Am J Physiol Endocrinol Metab ; 315(4): E705-E714, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29969314

ABSTRACT

Inflammatory diseases are associated with muscle wasting as a result of an increase in proteolysis. The purpose of this study was to elucidate whether administration of a ß2 adrenergic agonist, formoterol, was able to prevent the acute effects of sepsis induced by liposaccharide (LPS) injection on rat gastrocnemius muscle and to evaluate the possible roles of corticosterone, IGF-I, miR-23a, and miR-29b. For this purpose, male Wistar rats were injected with LPS and/or formoterol. Formoterol treatment decreased LPS-induced increase in serum corticosterone, TNFα upregulation, and NF-κB(p65) and Forkhead box protein O1 activation in the gastrocnemius. Atrogin-1, muscle RING-finger protein-1, microtubule-associated protein-1 light chain 3b (LC3b), and the lipidation of LC3b-I to LC3b-II were increased by LPS, and formoterol blocked these effects. Serum IGF-I and its mRNA levels in the gastrocnemius were decreased, whereas mecano growth factor and IGF binding protein 3 mRNA levels were increased in the rats injected with LPS but not in the rats that received LPS and formoterol. Similarly, LPS decreased Akt and mammalian target of rapamycin phosphorylation, and formoterol blocked these decreases. Finally, miR-29b expression in the gastrocnemius was upregulated by endotoxin injection, whereas miR-23a was not significantly different. Formoterol treatment did not significantly modify LPS-induced increase in muscle miR-29b. Furthermore, in control rats formoterol increased the expression of this miRNA. We conclude that formoterol decreases endotoxin-induced inflammation and proteolysis in rat skeletal muscle. Those responses can be a direct effect of ß2 adrenergic receptor stimulation or/and of blocking the effects of LPS on corticosterone and IGF-I. Muscle miR-23a and -29b do not seem to play an important role in those responses.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Formoterol Fumarate/pharmacology , Insulin-Like Growth Factor I/drug effects , Lipopolysaccharides/pharmacology , Muscle, Skeletal/drug effects , Muscular Atrophy/metabolism , Proteolysis/drug effects , Sepsis/metabolism , Animals , Corticosterone/metabolism , Insulin-Like Growth Factor I/metabolism , Male , MicroRNAs/drug effects , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
10.
Am J Obstet Gynecol ; 219(1): 113.e1-113.e9, 2018 07.
Article in English | MEDLINE | ID: mdl-29709511

ABSTRACT

BACKGROUND: Maternal inflammation is a risk factor for neonatal brain injury and future neurological deficits. Pomegranates have been shown to exhibit anti-inflammatory, anti-apoptotic and anti-oxidant activities. OBJECTIVE: We hypothesized that pomegranate juice (POM) may attenuate fetal brain injury in a rat model of maternal inflammation. STUDY DESIGN: Pregnant rats (24 total) were randomized for intraperitoneal lipopolysaccharide (100 µg/kg) or saline at time 0 at 18 days of gestation. From day 11 of gestation, 12 dams were provided ad libitum access to drinking water, and 12 dams were provided ad libitum access to drinking water with pomegranate juice (5 mL per day), resulting in 4 groups of 6 dams (saline/saline, pomegranate juice/saline, saline/lipopolysaccharide, pomegranate juice/lipopolysaccharide). All dams were sacrificed 4 hours following the injection and maternal blood and fetal brains were collected from the 4 treatment groups. Maternal interleukin-6 serum levels and fetal brain caspase 3 active form, nuclear factor-κB p65, neuronal nitric oxide synthase (phosphoneuronal nitric oxide synthase), and proinflammatory cytokine levels were determined by enzyme-linked immunosorbent assay and Western blot. RESULTS: Maternal lipopolysaccharide significantly increased maternal serum interleukin-6 levels (6039 ± 1039 vs 66 ± 46 pg/mL; P < .05) and fetal brain caspase 3 active form, nuclear factor-κB p65, phosphoneuronal nitric oxide synthase, and the proinflammatory cytokines compared to the control group (caspase 3 active form 0.26 ± 0.01 vs 0.20 ± 0.01 U; nuclear factor-κB p65 0.24 ± 0.01 vs 0.1 ± 0.01 U; phosphoneuronal nitric oxide synthase 0.23 ± 0.01 vs 0.11 ± 0.01 U; interleukin-6 0.25 ± 0.01 vs 0.09 ± 0.01 U; tumor necrosis factor-α 0.26 ± 0.01 vs 0.12 ± 0.01 U; chemokine (C-C motif) ligand 2 0.23 ± 0.01 vs 0.1 ± 0.01 U). Maternal supplementation of pomegranate juice to lipopolysaccharide-exposed dams (pomegranate juice/lipopolysaccharide) significantly reduced maternal serum interleukin-6 levels (3059 ± 1121 pg/mL, fetal brain: caspase 3 active form (0.2 ± 0.01 U), nuclear factor-κB p65 (0.22 ± 0.01 U), phosphoneuronal nitric oxide synthase (0.19 ± 0.01 U) as well as the brain proinflammatory cytokines (interleukin-6, tumor necrosis factor-α and chemokine [C-C motif] ligand 2) compared to lipopolysaccharide group. CONCLUSION: Maternal pomegranate juice supplementation may attenuate maternal inflammation-induced fetal brain injury. Pomegranate juice neuroprotective effects might be secondary to the suppression of both the maternal inflammatory response and inhibition of fetal brain apoptosis, neuronal nitric oxide synthase, and nuclear factor-κB activation.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Fetus/drug effects , Fruit and Vegetable Juices , Lipopolysaccharides/pharmacology , Lythraceae , Nitric Oxide Synthase Type I/drug effects , Transcription Factor RelA/drug effects , Animals , Antioxidants , Apoptosis/immunology , Brain/immunology , Brain/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Chemokine CCL2/drug effects , Chemokine CCL2/immunology , Cytokines/drug effects , Cytokines/immunology , Dietary Supplements , Female , Fetus/immunology , Fetus/metabolism , Inflammation , Interleukin-6/immunology , NF-kappa B/drug effects , NF-kappa B/immunology , NF-kappa B/metabolism , Nitric Oxide Synthase Type I/immunology , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Pregnancy , Rats , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology
11.
Rheumatology (Oxford) ; 56(8): 1417-1427, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28431044

ABSTRACT

Objective: To explore the role of leonurine in the regulation of synovial inflammation and joint destruction inRA. Methods: Fibroblast-like synoviocytes were isolated from synovial tissue from RA patients. Pro-inflammatory cytokine and MMP expression was evaluated using real-time PCR and a cytometric bead array. Cell migration and invasion in vitro were measured using the Boyden chamber method and the scratch assay, respectively. Protein expression was measured by western blotting. Nuclear factor kappa B (NF-κB) nuclear translocation was detected by immunofluorescence. The in vivo effect of leonurine was evaluated in mice with CIA. Results: Leonurine treatment significantly decreased the production of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8 and TNFα) and MMPs (MMP-1 and MMP-3) and suppressed the migration and invasion of RA fibroblast-like synoviocytes. The molecular analysis revealed that leonurine impaired TNFα-induced NF-κB signalling by inhibiting the phosphorylation and degradation of inhibitor of NF-κB alpha (IκBα) and subsequently preventing the nuclear translocation of the NF-κB p65 subunit. Leonurine also inhibited the p38 and Jun N-terminal kinase mitogen-activated protein kinases signalling pathways without affecting ERK signalling. Intraperitoneal injection of leonurine reduced synovial inflammation, joint destruction and the serum IL-1ß, IL-6 and TNFα levels in mice with CIA. Conclusion: Our findings show that leonurine reduces synovial inflammation and joint destruction in RA through the NF-κB and mitogen-activated protein kinases pathways. Leonurine has potential as a therapeutic agent for RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Gallic Acid/analogs & derivatives , Adult , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Cytokines/drug effects , Female , Fibroblasts/metabolism , Gallic Acid/pharmacokinetics , Humans , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 3/drug effects , Mice , Middle Aged , Mitogen-Activated Protein Kinases/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Synovial Membrane/drug effects , Synoviocytes/drug effects , Synoviocytes/pathology , Transcription Factor RelA/drug effects
12.
World J Urol ; 35(8): 1213-1221, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28105499

ABSTRACT

PURPOSE: The aberrant expression of casein kinase 2 (CK2) has been reported to be involved in the tumorigenesis and progression of prostate cancer. The inhibition of CK2 activity represses androgen-dependent prostate cancer cells by attenuating the androgen receptor (AR) signaling pathway. In this study, we examined the effect of CK2 inhibition in castration-resistant prostate cancer (CRPC) cells, in which AR variants (ARVs) play a predominant role. METHODS: A newly synthetic CK2 selective inhibitor CX4945 was utilized to study the effect of CK2 inhibition in CRPC cells by CCK8 assay and colony formation assay. Protein and mRNA levels of full-length AR (AR-FL) and AR-V7 were determined by qPCR and western blot, respectively. The nuclear translocation of p50 and p65 was assessed to reflect the activity of the NF-κB pathway. RESULTS: CX4945 reduced the proliferation of CRPC cells in a dose-dependent and time-dependent manner. AR-V7 rather than AR-FL was downregulated by CX4945 in both the mRNA and protein level. Furthermore, CX4945 could restore the sensitivity of CRPC cells to bicalutamide. The analysis of possible mechanisms demonstrated that the inhibition of CK2 diminished the phosphorylation of p65 at ser529 and thus attenuated the activity of the NF-κB pathway. CONCLUSION: The inhibition of CK2 by CX4945 can repress the viability of CRPC cells and restore their sensitivity to anti-androgen therapy by suppressing AR-V7. This finding presents a potential option for the treatment of prostate cancer, especially CRPC.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Naphthyridines/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/drug effects , Androgen Antagonists/pharmacology , Anilides/pharmacology , Blotting, Western , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Genetic Variation , Humans , Male , NF-kappa B/drug effects , NF-kappa B/metabolism , NF-kappa B p50 Subunit/drug effects , NF-kappa B p50 Subunit/metabolism , Nitriles/pharmacology , Phenazines , Prostatic Neoplasms, Castration-Resistant/genetics , RNA, Messenger/drug effects , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction , Tosyl Compounds/pharmacology , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , Tumor Stem Cell Assay
13.
Am J Physiol Heart Circ Physiol ; 311(4): H871-H880, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27496879

ABSTRACT

We previously reported that endoplasmic reticulum (ER) stress is induced in the subfornical organ (SFO) and the hypothalamic paraventricular nucleus (PVN) of heart failure (HF) rats and is reduced by inhibition of mitogen-activated protein kinase (MAPK) signaling. The present study further examined the relationship between brain MAPK signaling, ER stress, and sympathetic excitation in HF. Sham-operated (Sham) and HF rats received a 4-wk intracerebroventricular (ICV) infusion of vehicle (Veh) or the ER stress inhibitor tauroursodeoxycholic acid (TUDCA, 10 µg/day). Lower mRNA levels of the ER stress biomarkers GRP78, ATF6, ATF4, and XBP-1s in the SFO and PVN of TUDCA-treated HF rats validated the efficacy of the TUDCA dose. The elevated levels of phosphorylated p44/42 and p38 MAPK in SFO and PVN of Veh-treated HF rats, compared with Sham rats, were significantly reduced in TUDCA-treated HF rats as shown by Western blot and immunofluorescent staining. Plasma norepinephrine levels were higher in Veh-treated HF rats, compared with Veh-treated Sham rats, and were significantly lower in the TUDCA-treated HF rats. TUDCA-treated HF rats also had lower mRNA levels for angiotensin converting enzyme, angiotensin II type 1 receptor, tumor necrosis factor-α, interleukin-1ß, cyclooxygenase-2, and NF-κB p65, and a higher mRNA level of IκB-α, in the SFO and PVN than Veh-treated HF rats. These data suggest that ER stress contributes to the augmented sympathetic activity in HF by inducing MAPK signaling, thereby promoting inflammation and renin-angiotensin system activity in key cardiovascular regulatory regions of the brain.


Subject(s)
Brain/metabolism , Endoplasmic Reticulum Stress , Heart Failure/metabolism , Inflammation/metabolism , Mitogen-Activated Protein Kinases/metabolism , Renin-Angiotensin System , Sympathetic Nervous System/metabolism , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/genetics , Activating Transcription Factor 6/drug effects , Activating Transcription Factor 6/genetics , Animals , Blotting, Western , Brain/drug effects , Cholagogues and Choleretics/pharmacology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Echocardiography , Heart Failure/physiopathology , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Infusions, Intraventricular , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Male , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/drug effects , NF-KappaB Inhibitor alpha/drug effects , NF-KappaB Inhibitor alpha/genetics , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Peptidyl-Dipeptidase A/drug effects , Peptidyl-Dipeptidase A/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Signal Transduction , Subfornical Organ/drug effects , Subfornical Organ/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiopathology , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , X-Box Binding Protein 1/drug effects , X-Box Binding Protein 1/genetics , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Osteoarthritis Cartilage ; 24(1): 178-187, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26256766

ABSTRACT

OBJECTIVE: To determine whether mandibular condylar cartilage degradation induced by experimentally abnormal occlusion could be ameliorated via systemic administration of strontium or NBD peptide. METHODS: Six-week-old female C57BL/6J mice were used. From the seventh day after mock operation or unilateral anterior crossbite (UAC) treatment, the control and UAC mice were further respectively pharmacologically treated for 2 weeks or 4 weeks of saline (CON + Saline and UAC + Saline groups), SrCl2 (CON + SrCl2 and UAC + SrCl2 groups) or NBD peptide (CON + NBD peptide and UAC + NBD peptide groups). Changes in condylar cartilage and subchondral bone were assessed 21 and 35 days after mock operation or UAC procedure by histology and micro-CT. Real-time PCR and/or immunohistochemistry (IHC) were performed to evaluate changes in expression levels of col2a1, aggrecan, ADAMTS-5, tnf-α, il-1ß, nfkbia, nuclear factor-kappaB phospho-p65 in condylar cartilage, and rankl/rank/opg in both condylar cartilage and subchondral bone. RESULTS: Cartilage degradation with decreased col2a1 and aggrecan expression, and increased ADAMTS-5, tnf-α/il1-ß, nfkbia and NF-κB phospho-p65 was observed in UAC + Saline groups. Subchondral bone loss with increased osteoclast numbers and decreased opg/rankl ratio was found in UAC + Saline groups compared to age-match CON + Saline groups. Cartilage degradation and subchondral bone loss were reversed by treatment of SrCl2 or NBD peptide while the same dosage in control mice induced few changes in condylar cartilage and subchondral bone. CONCLUSIONS: The results demonstrate reverse effect of systemic administration of strontium or NBD peptide on UAC-induced condylar cartilage degradation and subchondral bone loss.


Subject(s)
Cartilage, Articular/drug effects , Malocclusion , Mandibular Condyle/drug effects , Osteoclasts/drug effects , Peptides/pharmacology , RNA, Messenger/drug effects , Strontium/pharmacology , ADAM Proteins/drug effects , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS5 Protein , Aggrecans/drug effects , Aggrecans/genetics , Aggrecans/metabolism , Animals , Cartilage, Articular/metabolism , Collagen Type II/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Dental Occlusion , Female , I-kappa B Proteins/drug effects , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Immunohistochemistry , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mandibular Condyle/metabolism , Mandibular Condyle/pathology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Osteoclasts/metabolism , Osteoprotegerin/drug effects , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Proteoglycans/drug effects , Proteoglycans/metabolism , RANK Ligand/drug effects , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor Activator of Nuclear Factor-kappa B/drug effects , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
J Nat Prod ; 79(4): 1105-11, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27019105

ABSTRACT

A new secondary metabolite, named griseusrazin A (1), was isolated from the marine-derived bacterium Streptomyces griseus subsp. griseus. The structure of the compound was determined by analysis of spectroscopic data including MS, COSY, HSQC, HMBC, and (15)N-HMBC data. Griseusrazin A (1) inhibited the production of inflammatory mediators, such as prostaglandin E2 and nitric oxide, which was mediated through the suppression of the expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. The production of pro-inflammatory cytokines, such as interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α, in the LPS-stimulated cells was also effectively blocked by griseusrazin A (1). Furthermore, this anti-inflammatory activity of 1 was linked to its inhibitory effects against the nuclear translocation of NF-κB p50 and p65, as wells as NF-κB binding activity. In the further study to elucidate the anti-inflammatory mechanism, 1 was shown to induce heme oxygenase-1 (HO-1) expression through the enhancement of nuclear translocation of nuclear factor E2-related factor 2. Furthermore, the anti-inflammatory activity of 1 in the LPS-stimulated cells was partially reversed by an HO inhibitor, tin protoporphyrin. These results indicate that the anti-inflammatory effect of 1 is associated with Nrf2-mediated HO-1 expression.


Subject(s)
Heme Oxygenase-1/drug effects , Pyrazines/pharmacology , Animals , Cytokines/drug effects , Dose-Response Relationship, Drug , Interleukin-1beta/drug effects , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Marine Biology , Mice , Molecular Structure , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/drug effects , Nuclear Magnetic Resonance, Biomolecular , Pyrazines/chemistry , Pyrazines/isolation & purification , RNA, Messenger/drug effects , Streptomyces griseus/chemistry , Transcription Factor RelA/drug effects , Tumor Necrosis Factor-alpha/drug effects
16.
Clin Immunol ; 159(1): 13-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25926428

ABSTRACT

We showed previously that 17ß estradiol (E2) led to improved survival in nephrotoxic serum induced nephritis (NTN) in male mice. In this study we determined whether E2 regulates vascular cell adhesion molecule (VCAM)-1, an adhesion molecule that is upregulated in kidney during autoimmune nephritis, in mesangial cells (MC). We show that E2 inhibited VCAM-1 up-regulation in kidneys in vivo during NTN, and in MCs upon TNFα stimulation. VCAM-1 up-regulation in MCs was controlled by the transcription factor NFκB. E2 inhibited RNA polymerase II recruitment to the VCAM-1 promoter, but not p65 recruitment. Interestingly E2 inhibited TNFα stimulated interaction between poly (ADP-ribose) polymerase-1 (PARP-1) and p65. As PARP-1 is required for VCAM-1 upregulation in MCs, our data suggest that E2 may inhibit pre-initiation complex formation at VCAM-1 promoter by inhibiting PARP-1 recruitment to p65. We propose that E2 plays an important role in regulating renal inflammation locally.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Glomerulonephritis , Mesangial Cells/drug effects , RNA, Messenger/metabolism , Vascular Cell Adhesion Molecule-1/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Gene Expression/drug effects , Kidney/drug effects , Kidney/metabolism , Mesangial Cells/metabolism , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , RNA Polymerase II/drug effects , RNA Polymerase II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , Up-Regulation , Vascular Cell Adhesion Molecule-1/genetics
17.
J Pharmacol Exp Ther ; 353(3): 539-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25862641

ABSTRACT

Endotoxin-induced acute inflammatory diseases such as sepsis, mediated by excessive production of various proinflammatory cytokines, remain the leading cause of mortality in critically ill patients. Lipopolysaccharide (LPS), the characteristic endotoxin found in the outer membrane of Gram-negative bacteria, can induce the innate immunity system and through the myeloid differentiation protein 2 (MD2) and Toll-like receptor 4 (TLR4) complex, increase the production of inflammatory mediators. Our previous studies have found that a curcumin analog, L48H37 [1-ethyl-3,5-bis(3,4,5-trimethoxybenzylidene)piperidin-4-one], was able to inhibit LPS-induced inflammation, particularly tumor necrosis factor α and interleukin 6 production and gene expression in mouse macrophages. In this study, a series of biochemical experiments demonstrate L48H37 specifically targets MD2 and inhibits the interaction and signaling transduction of LPS-TLR4/MD2. L48H37 binds to the hydrophobic region of MD2 pocket and forms hydrogen bond interactions with Arg(90) and Tyr(102). Subsequently, L48H37 was shown to suppress LPS-induced mitogen-activated protein kinase phosphorylation and nuclear factor κB activation in macrophages; it also dose dependently inhibits the cytokine expression in macrophages and human peripheral blood mononuclear cells stimulated by LPS. In LPS-induced septic mice, both pretreatment and treatment with L48H37 significantly improved survival and protected lung injury. Taken together, this work identified a new MD2 specific inhibitor, L48H37, as a potential candidate in the treatment of sepsis.


Subject(s)
Curcumin/analogs & derivatives , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/antagonists & inhibitors , Sepsis/drug therapy , Toll-Like Receptor 4/drug effects , Animals , Curcumin/metabolism , Curcumin/pharmacology , Cytokines/biosynthesis , Diarylheptanoids , Endotoxins/toxicity , Humans , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred C57BL , Primary Cell Culture , Protein Binding/drug effects , Shock, Septic/drug therapy , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/drug effects
18.
Histochem Cell Biol ; 144(3): 223-35, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25990946

ABSTRACT

To evaluate the role of actin cytoskeleton in the regulation of NF-κB transcription factor, we analyzed its involvement in the intracellular transport and nuclear translocation of the NF-κB RelA/p65 subunit in A431 epithelial cells stimulated with fibronectin and EGF. Live cell imaging and confocal microscopy showed that EGF activated the movement of RelA/p65 in the cytoplasm. Upon cell adhesion to fibronectin, RelA/p65 concentrated onto stress fibers, and EGF stimulated its subsequent allocation to membrane ruffles, newly organized stress fibers, and discrete cytoplasmic actin-rich patches. These patches also contained α-actinin-1 and α-actinin-4, vinculin, paxillin, α-tubulin, and PI3-kinase. Cytochalasin D treatment resulted in RelA/p65 redistribution to actin-containing aggregates, with the number of cells with RelA/p65-containing clusters in the cytoplasm increasing under the effect of EGF. Furthermore, EGF proved to induce RelA/p65 accumulation in the nucleus after cell pretreatment with actin-stabilizing and actin-destabilizing agents, which was accompanied by changes in its DNA-binding activity after either EGF stimulation or cytochalasin D treatment. Thus, EGF treatment of A431 cells results in simultaneous nuclear RelA/p65 translocation and cytoplasmic redistribution, with part of RelA/p65 pool forming a very tight association with actin-rich structures. Apparently, nuclear transport is independent on drug stabilization or destabilization of the actin.


Subject(s)
Actins/metabolism , Epidermal Growth Factor/pharmacology , Fibronectins/metabolism , Transcription Factor RelA/metabolism , Actins/drug effects , Cell Line , Cell Line, Tumor , Cytochalasin D/pharmacology , Cytoplasm/drug effects , Cytoplasm/metabolism , Humans , NF-kappa B/biosynthesis , Nucleic Acid Synthesis Inhibitors/pharmacology , Transcription Factor RelA/drug effects , Translocation, Genetic/drug effects
19.
Drug Metab Dispos ; 43(8): 1181-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986850

ABSTRACT

Pregnane X receptor (PXR) activation exhibits anti-inflammatory effects via repressing nuclear factor-κB (NF-κB); however, its overactivation may disrupt homeostasis of various enzymes and transporters. Here we found that ginsenosides restore PXR/NF-κB signaling in inflamed conditions without disrupting PXR function in normal conditions. The effects and mechanisms of ginsenosides in regulating PXR/NF-κB signals were determined both in vitro and in vivo. Ginsenosides significantly inhibited NF-κB activation and restored the expression of PXR target genes in tumor necrosis factor-α-stimulated LS174T cells. Despite not being PXR agonists, ginsenosides repressed NF-κB activation in a PXR-dependent manner. Ginsenosides significantly increased the physical association between PXR and the NF-κB p65 subunit and thereby decreased the nuclear translocation of p65. Ginsenoside Rb1 and compound K (CK) were major bioactive compounds in the regulating PXR/NF-κB signaling. Consistently, ginsenosides significantly attenuated dextran sulfate sodium-induced experimental colitis, which was associated with restored PXR/NF-κB signaling. This study indicates that ginsenosides may elicit anti-inflammatory effects via targeting PXR/NF-κB interaction without disrupting PXR function in healthy conditions. Ginsenoside Rb1 and CK may serve as leading compounds in the discovery of new drugs that target PXR/NF-κB interaction in therapy for inflammatory bowel disease.


Subject(s)
Colitis/drug therapy , Ginsenosides/pharmacology , NF-kappa B/drug effects , Neuroprotective Agents/pharmacology , Receptors, Steroid/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Ginsenosides/therapeutic use , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Pregnane X Receptor , Signal Transduction/drug effects , Transcription Factor RelA/drug effects , Tumor Necrosis Factor-alpha/pharmacology
20.
BMC Nephrol ; 16: 72, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25975951

ABSTRACT

BACKGROUND: B cell activating factor belonging to the TNF family (BAFF) is vital for B cell survival, proliferation and activation. Evidence indicates that BAFF is systemically or locally increased in glomerulonephritis (e.g. lupus nephritis, IgA nephropathy). However, the effect of BAFF on human mesangial cells is not known. METHODS: The impact of BAFF on the proliferation of a human mesangial cell line in vitro was investigated. The expression of BAFF receptor (BAFF-R) and downstream signal transduction were explored. The influence of BAFF on the expression of related genes was also studied. RESULTS: Our data indicated that BAFF had a proliferative effect on human mesangial cells, as supported by the results of cell proliferation assays and the inhibited expression of the pro-apoptotic gene Bim. BAFF-R was expressed on the cell membrane of human mesangial cells and blockade of BAFF/BAFF-R binding abrogated the proliferative effect of BAFF on human mesangial cells. BAFF stimulation led to rapid phosphorylation of NF-κBp65, Akt and MAPK p38 kinase in human mesangial cells, whereas it had no effect on the expression of NF-κB p100 and phosphorylation of Erk. The phosphorylation of Akt was very sensitive to blockade of BAFF/BAFF-R ligation, although activation of MAPK p38 and NF-κBp65 was not. BAFF treatment resulted in decreased expression of BAFF-R, which implied negative feedback regulation after its binding. CONCLUSIONS: BAFF promoted proliferation of human mesangial cells, which was mediated via BAFF-R. The BAFF/BAFF-R interaction triggered Akt, p65 and p38 activation, with Akt phosphorylation being tightly dependent on BAFF/BAFF-R interaction.


Subject(s)
B-Cell Activating Factor/pharmacology , B-Cell Activation Factor Receptor/drug effects , Cell Proliferation/drug effects , Mesangial Cells/drug effects , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , B-Cell Activation Factor Receptor/metabolism , Bcl-2-Like Protein 11 , Cells, Cultured , Humans , In Vitro Techniques , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mesangial Cells/metabolism , NF-kappa B p52 Subunit/drug effects , NF-kappa B p52 Subunit/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL