Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
J Pharmacol Sci ; 154(4): 225-235, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38485340

ABSTRACT

In this study, we investigated the regulatory mechanisms underlying the effects of LPS tolerance on the inflammatory homeostasis of immune cells. LPS priming-induced immune tolerance downregulated cyclooxygenase-2, and lowered the production of prostaglandin-E2 in microglial cells. In addition, LPS tolerance downregulated the expression of suppressor of cytokine signaling 3, and inducible nitric oxide synthase/nitric oxide; suppressed the LPS-mediated induction of tumor necrosis factor-α, interleukin (IL)-6, and IL-1; and reduced reactive oxygen species production in microglial cells. LPS stimulation increased the levels of the adaptive response-related proteins heme oxygenase-1 and superoxide dismutase 2, and the levels of heme oxygenase-1 (HO-1) enhanced after LPS priming. Systemic administration of low-dose LPS (0.5 mg/kg) to mice for 4 consecutive days attenuated high-dose LPS (5 mg/kg)-induced inflammatory response, microglial activation, and proinflammatory cytokine expression. Moreover, repeated exposure to low-dose LPS suppressed the recruitment of peripheral monocytes or macrophages to brain regions and downregulated the expression of proinflammatory cytokines. Notably, LPS-induced social avoidance behaviors in mice were mitigated by immune tolerance. In conclusion, immune tolerance may reduce proinflammatory cytokine expression and reactive oxygen species production. Our findings provide insights into the effects of endotoxin tolerance on innate immune cells and social behaviors.


Subject(s)
Heme Oxygenase-1 , Microglia , Animals , Mice , Heme Oxygenase-1/metabolism , Microglia/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Avoidance Learning , Cytokines/metabolism , Interleukin-6/metabolism , Social Behavior , Immune Tolerance , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism
2.
Environ Toxicol ; 36(12): 2551-2561, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34520103

ABSTRACT

Cancer and its associated conditions have significant impacts on public health at many levels worldwide, and cancer is the leading cause of death among adults. Peroxisome proliferator-activated receptor α (PPARα)-specific agonists, fibrates, have been approved by the Food and Drug Administration for managing hyperlipidemia. PPARα-specific agonists exert anti-cancer effects in many human cancer types, including glioblastoma (GBM). Recently, we have reported that the hypoxic state in GBM stabilizes hypoxia-inducible factor-1 alpha (HIF-1α), thus contributing to tumor escape from immune surveillance by activating the expression of the pH-regulating protein carbonic anhydrase IX (CA9). In this study, we aimed to study the regulatory effects of the PPARα agonist fibrate on the regulation of HIF-1α expression and its downstream target protein in GBM. Our findings showed that fenofibrate is the high potency compound among the various fibrates that inhibit hypoxia-induced HIF-1α and CA9 expression in GBM. Moreover, fenofibrate-inhibited HIF-1α expression is mediated by HO-1 activation in GBM cells through the AMP-activated protein kinase (AMPK) pathway. In addition, fenofibrate-enhanced HO-1 upregulation activates SIRT1 and leads to subsequent accumulation of SIRT1 in the nucleus, which further promotes HIF-1α deacetylation and inhibits CA9 expression. Using a protein synthesis inhibitor, cycloheximide, we also observed that fenofibrate inhibited HIF-1α protein synthesis. In addition, the administration of the proteasome inhibitor MG132 showed that fenofibrate promoted HIF-1α protein degradation in GBM. Hence, our results indicate that fenofibrate is a useful anti-GBM agent that modulates hypoxia-induced HIF-1α expression through multiple cellular pathways.


Subject(s)
Carbonic Anhydrases , Fenofibrate , Glioblastoma , AMP-Activated Protein Kinases/genetics , Fenofibrate/pharmacology , Glioblastoma/genetics , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Sirtuin 1
3.
Molecules ; 26(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34684832

ABSTRACT

Androgens have been shown to have a beneficial effect on brain injury and lower reactive astrocyte expression after TBI. Androgen receptors (ARs) are known to mediate the neuroprotective effects of androgens. However, whether ARs play a crucial role in TBI remains unknown. In this study, we investigated the role of ARs in TBI pathophysiology, using AR knockout (ARKO) mice. We used the controlled cortical impact model to produce primary and mechanical brain injuries and assessed motor function and brain-lesion volume. In addition, the AR knockout effects on necrosis and autophagy were evaluated after TBI. AR knockout significantly increased TBI-induced expression of the necrosis marker alpha-II-spectrin breakdown product 150 and astrogliosis marker glial fibrillary acidic protein. In addition, the TBI-induced astrogliosis increase in ARKO mice lasted for three weeks after a TBI. The autophagy marker Beclin-1 was also enhanced in ARKO mice compared with wild-type mice after TBI. Our results also indicated that ARKO mice showed a more unsatisfactory performance than wild-type mice in a motor function test following TBI. Further, they were observed to have more severe lesions than wild-type mice after injury. These findings strongly suggest that ARs play a role in TBI.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Receptors, Androgen/deficiency , Animals , Autophagy , Beclin-1/metabolism , Brain/physiology , Brain/physiopathology , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mice , Mice, Knockout , Motor Disorders/pathology , Motor Disorders/physiopathology , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Spectrin/metabolism
4.
Int J Mol Sci ; 21(16)2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32823915

ABSTRACT

Carbonic anhydrases (CAs) are acid-base regulatory proteins that modulate a variety of physiological functions. Recent findings have shown that CAIX is particularly upregulated in glioblastoma multiforme (GBM) and is associated with a poor patient outcome and survival rate. An analysis of the GSE4290 dataset of patients with gliomas showed that CAIX was highly expressed in GBM and was negatively associated with prognosis. The expression of CAIX under hypoxic conditions in GBM significantly increased in protein, mRNA, and transcriptional activity. Importantly, CAIX upregulation also regulated GBM motility, monocyte adhesion to GBM, and the polarization of tumor-associated monocytes/macrophages (TAM). Furthermore, the overexpression of CAIX was observed in intracranial GBM cells. Additionally, epidermal growth factor receptor/signal transducer and activator of transcription 3 regulated CAIX expression under hypoxic conditions by affecting the stability of hypoxia-inducible factor 1α. In contrast, the knockdown of CAIX dramatically abrogated the change in GBM motility and monocyte adhesion to GBM under hypoxic conditions. Our results provide a comprehensive understanding of the mechanisms of CAIX in the GBM microenvironment. Hence, novel therapeutic targets of GBM progression are possibly developed.


Subject(s)
Carbonic Anhydrase IX/metabolism , Cell Movement , ErbB Receptors/metabolism , Glioblastoma/enzymology , Glioblastoma/pathology , STAT3 Transcription Factor/metabolism , Tumor Hypoxia , Tumor-Associated Macrophages/pathology , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Polarity , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Monocytes/pathology , Tumor Microenvironment , Tumor-Associated Macrophages/enzymology
5.
J Proteome Res ; 18(1): 182-190, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30351951

ABSTRACT

Stressful events promote psychopathogenic changes that might contribute to the development of mental illnesses. Some individuals tend to recover from the stress response, while some do not. However, the molecular mechanisms of stress resilience during stress are not well-characterized. Here, we identify proteomic changes in the hippocampus using proteomic technique to examine mice following chronic social defeat stress. We showed that small ubiquitin-like modifier (SUMO)-1 expression was significantly decreased in susceptible mice following chronic social defeat stress. We also examined a protein inhibitor of activated signal transducer of transcription (PIAS)1 levels, an E3 SUMO-protein ligase protein inhibitor of activated STAT1, which is known to interact with SUMO-1. PIAS1 was shown to be profoundly decreased and monoamine oxidase (MAO)-A increased in the hippocampus of susceptible mice following chronic social defeat stress. Furthermore, the manipulated PIAS1 expression in the hippocampus also has an influence on glucocorticoid receptor (GR) translocation. We also found that knockdown of PIAS1 expression in the hippocampus then subject to submaximal stress increased GR to glucocorticoid response element (GRE)-binding site on the MAO-A promoter. The present study raises the possibility of different levels of PIAS1 between individuals in response to chronic social defeat stress and that such differences may contribute to the susceptibility to stress.


Subject(s)
Protein Inhibitors of Activated STAT/metabolism , Proteolysis , Proteomics/methods , Stress, Psychological/metabolism , Animals , Chronic Disease , Hippocampus/metabolism , Mice , Small Ubiquitin-Related Modifier Proteins/metabolism
6.
Toxicol Appl Pharmacol ; 338: 182-190, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180066

ABSTRACT

Connexins are widely supported as tumor suppressors due to their downregulation in cancers, nevertheless, more recent evidence suggests roles for connexins in facilitating tumor progression in later stages, including metastasis. One of the key factors regulating the expression, modification, stability, and localization of connexins is hormone receptors in hormone-dependent cancers. It is reasonable to consider that hormones/hormone receptors may modulate connexins expression and play critical roles in the cellular control of connexins during breast cancer progression. In estrogen receptor (ER)-positive breast cancers, tamoxifen and fulvestrant are widely used therapeutic agents and are considered to alter ER signaling. In this present study, we investigated the effects of fulvestrant and tamoxifen in Cx43 expression, and we also explored the role of Cx43 in ER-positive breast cancer migration and the relationship between Cx43 and ER. The involvement of estrogen/ER in Cx43 modulation was further verified by administering tyrosine kinase inhibitors and chemotherapeutic agents. We found that inhibition of ER promoted the binding of E3 ligase Nedd4 to Cx43, leading to Cx43 ubiquitination. Furthermore, inhibition of ER by fulvestrant and tamoxifen phosphorylated p38 MAPK, and inhibition of Rac, MKK3/6, and p38 reversed fulvestrant-reduced Cx43 expression. These findings suggest that Cx43 expression which may positively regulate cell migration is ER-dependent in ER-positive breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Connexin 43/physiology , Estrogen Antagonists/pharmacology , Breast Neoplasms/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Connexin 43/analysis , Female , Humans , Nedd4 Ubiquitin Protein Ligases/metabolism , Receptors, Estrogen/physiology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology
7.
Int J Mol Sci ; 20(1)2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30585203

ABSTRACT

OBJECTIVE: Intervertebral disc (IVD) degeneration and disc herniation are major causes of lower back pain, which involve the presence of inflammatory mediators and tissue invasion by immune cells. Intercellular adhesion molecule 1 (ICAM1, also termed CD54) is an adhesion molecule that mediates cell-cell interactions, particularly between immune cells and target tissue. The aim of this study was to examine the intracellular signaling pathways involved in inflammatory stimuli-induced ICAM1 expression in human anulus fibrosus (AF) cells. METHODS: Quantitative reverse transcription-polymerase chain reaction (qPCR), western blotting, and flow cytometry were performed to dissect the roles of different signaling pathways in inflammatory stimuli-mediated ICAM1 expression. RESULTS: Using qPCR and western blot analyses, a significant increase in ICAM1 expression was observed in AF cells after stimulation of lipopolysaccharide (LPS) plus interferon-gamma (IFNγ) in a time-dependent manner. Flow cytometry revealed ICAM1 upregulation on the surface of AF cells. Importantly, LPS plus IFNγ treatment also significantly promoted Chemokine ligand (CCL)2 expression, but not CCL3. The enhanced ICAM1 expression was abolished after incubation with antibody against CCL2. In AF cells, treatment with LPS plus IFNγ activated the FAK/ERK/GSK3 signaling pathways, promoted a time-dependent increase in PKCδ phosphorylation, and promoted PKCδ translocation to the nucleus. Treatment with the pharmacological PKCδ inhibitor; rottlerin, effectively blocked the enhanced productions of ICAM1 and CCL2. CONCLUSIONS: Inflammatory stimuli in AF cells are part of a specific pathophysiology in IVD degeneration and disc herniation that modulates CCL2/ICAM1 activation through the FAK/ERK/GSK3 and PKCδ signaling pathways in AF cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Glycogen Synthase Kinase 3/metabolism , Intercellular Adhesion Molecule-1/metabolism , Protein Kinase C-delta/metabolism , Acetophenones/pharmacology , Annulus Fibrosus/cytology , Annulus Fibrosus/metabolism , Benzopyrans/pharmacology , Chemokine CCL2/metabolism , Humans , Interferon-gamma/pharmacology , Janus Kinase 2/metabolism , Lipopolysaccharides/pharmacology , Phosphorylation/drug effects , Protein Kinase C-delta/antagonists & inhibitors , Signal Transduction/drug effects , Up-Regulation/drug effects
8.
Int J Mol Sci ; 19(1)2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29301329

ABSTRACT

Glioblastoma multiforme (GBM) is the most common type of primary and malignant tumor occurring in the adult central nervous system. Temozolomide (TMZ) has been considered to be one of the most effective chemotherapeutic agents to prolong the survival of patients with glioblastoma. Many glioma cells develop drug-resistance against TMZ that is mediated by increasing O-6-methylguanine-DNA methyltransferase (MGMT) levels. The expression of connexin 43 was increased in the resistant U251 subline compared with the parental U251 cells. The expression of epithelial-mesenchymal transition (EMT)-associated regulators, including vimentin, N-cadherin, and ß-catenin, was reduced in the resistant U251 subline. In addition, the resistant U251 subline exhibited decreased cell migratory activity and monocyte adhesion ability compared to the parental U251 cells. Furthermore, the resistant U251 subline also expressed lower levels of vascular cell adhesion molecule (VCAM)-1 after treatment with recombinant tumor necrosis factor (TNF)-α. These findings suggest differential characteristics in the drug-resistant GBM from the parental glioma cells.


Subject(s)
Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioma/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Connexin 43/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Humans , Monocytes/drug effects , Monocytes/pathology , Temozolomide , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , Vascular Cell Adhesion Molecule-1/metabolism
9.
J Mol Cell Cardiol ; 99: 113-122, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27503317

ABSTRACT

Lumbrokinase, a novel antithrombotic agent, purified from the earthworm Lumbricus rubellus, has been clinically used to treat stroke and cardiovascular diseases. However, inflammatory responses associated with the cardioprotective effect of lumbrokinase remain unknown. In this study, the signaling pathways involved in lumbrokinase-inhibited expressions of inflammation mediators were investigated in rats subjected to myocardial ischemia-reperfusion (I-R) injury. The left main coronary artery of anesthetized rats was subjected to 1h occlusion and 3h reperfusion. The animals were treated with/without lumbrokinase and the severities of I-R-induced arrhythmias and infarction were compared. Lumbrokinase inhibited I-R-induced arrhythmias and reduced mortality, as well as decreased the lactate dehydrogenase levels in carotid blood. Lumbrokinase also inhibited the enhancement of I-R induced expressions of cyclooxygenase (COX)-2, inducible nitric oxide synthase (iNOS), and matrix metalloproteinase (MMP)-9 through toll-like receptor 4 (TLR4) signaling pathway. Moreover, our results demonstrated that stimulation with lumbrokinase decreases the phosphorylation of JNK, IκB, and NF-κB. These findings suggested that lumbrokinase is a potent cardioprotective drug in rats with I-R injury. The cardioprotective effects of lumbrokinase may be correlated with its inhibitory effect on the I-R-induced expressions of COX-2, iNOS and MMP-9, mediated by TLR4 signaling through JNK and NF-κB pathways.


Subject(s)
Biological Products/pharmacology , Endopeptidases/pharmacology , Myocardial Reperfusion Injury/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Biomarkers , Cyclooxygenase 2/metabolism , Electrocardiography , Heart Rate , Hemodynamics , Male , Matrix Metalloproteinases/metabolism , Myocardial Infarction/diagnosis , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/diagnosis , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase/metabolism , Peroxidase/metabolism , Rats , Toll-Like Receptor 2/metabolism
10.
J Neuroinflammation ; 13: 21, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26819091

ABSTRACT

BACKGROUND: Alzheimer's disease is associated with amyloid-beta (Aß)-induced microglia activation. This pro-inflammatory response promotes neuronal damage, and therapies are sought to limit microglial activation. Screening efforts to develop new pharmacological inhibitors require a robust in vitro cell system. Current models lack significant responses to Aß, and their use in examining age-related neurodegenerative diseases is questionable. For example, the commonly used BV-2 microglial line was derived from embryonic mononuclear cells and its activation by various stimuli is limited. To this end, we have established a new immortalized microglial (IMG) cell line from adult murine brain. The objective of this study was to characterize Aß-induced activation of IMG cells, and here, we demonstrate the ability of cannabinoids to significantly reduce this inflammatory response. METHODS: Microglial cells derived from adult murine brain were immortalized via infection with the v-raf/v-myc retrovirus under conditions that selectively promote microglia growth. The presence or absence of markers CD11b and F4/80 (microglial), NeuN (neuronal), and GFAP (astrocytic) was assessed by immunofluorescence microscopy and western blotting. Using IMG and BV-2 cells, levels of pro- and anti-inflammatory transcripts in response to extracellular stimuli were determined by quantitative PCR (qPCR). Phagocytosis of fluorescent beads and fluorescein isothiocyanate (FITC)-labeled Aß oligomers was assessed using flow cytometry and fluorescence microscopy. FITC-Aß uptake was quantified using a fluorescence plate reader. The ability of cannabinoids to mitigate Aß-induced expression of inducible nitric oxide synthase (iNOS) was evaluated. RESULTS: IMG cells express the microglial markers CD11b and F4/80 but not NeuN or GFAP. Relative to BV-2 cells, IMG cells increased iNOS (>200-fold) and Arg-1 (>100-fold) in response to pro- and anti-inflammatory stimuli. IMG cells phagocytose foreign particles and Aß oligomers, with the latter trafficked to phagolysosomes. Aß-induced activation of IMG cells was suppressed by delta-9-tetrahydrocannabinol and the CB2-selective agonist JWH-015 in a time- and concentration-dependent manner. CONCLUSIONS: IMG cells recapitulate key features of microglial cell activation. As an example of their potential pharmacological use, cannabinoids were shown to reduce activation of Aß-induced iNOS gene expression. IMG cells hold promising potential for drug screening, mechanistic studies, and functional investigations directed towards understanding how Aß interacts with microglia.


Subject(s)
Amyloid beta-Peptides/metabolism , Microglia/metabolism , Analysis of Variance , Animals , Antigens, Differentiation/metabolism , CD11b Antigen/metabolism , Cells, Cultured , Flow Cytometry , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , In Vitro Techniques , Interleukin-1beta/metabolism , Microglia/drug effects , Phagocytes/metabolism , RNA, Messenger/metabolism
11.
Int J Mol Sci ; 16(4): 8844-60, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25906473

ABSTRACT

Increasing studies suggest that inflammatory processes in the central nervous system mediated by microglial activation plays an important role in numerous neurodegenerative diseases. Development of planning for microglial suppression is considered a key strategy in the search for neuroprotection. Paeonol is a major phenolic component of Moutan Cortex, widely used as a nutrient supplement in Chinese medicine. In this study, we investigated the effects of paeonol on microglial cells stimulated by inflammagens. Paeonol significantly inhibited the release of nitric oxide (NO) and the expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Treatment with paeonol also reduced reactive oxygen species (ROS) production and inhibited an ATP-induced increased cell migratory activity. Furthermore, the inhibitory effects of neuroinflammation by paeonol were found to be regulated by phosphorylated adenosine monophosphate-activated protein kinase-α (AMPK-α) and glycogen synthase kinase 3 α/ß (GSK 3α/ß). Treatment with AMPK or GSK3 inhibitors reverse the inhibitory effect of neuroinflammation by paeonol in microglial cells. Furthermore, paeonol treatment also showed significant improvement in the rotarod performance and microglial activation in the mouse model as well. The present study is the first to report a novel inhibitory role of paeonol on neuroinflammation, and presents a new candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Subject(s)
Acetophenones/pharmacology , Anti-Inflammatory Agents/pharmacology , Microglia/drug effects , Adenylate Kinase/metabolism , Animals , Cell Line , Cell Movement , Cells, Cultured , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Lipopolysaccharides/pharmacology , Male , Mice, Inbred ICR , Microglia/immunology , Motor Activity/drug effects , Signal Transduction
12.
Int J Mol Sci ; 16(3): 5572-89, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25768341

ABSTRACT

Microglial activation has been widely demonstrated to mediate inflammatory processes that are crucial in several neurodegenerative disorders. Pharmaceuticals that can deliver direct inhibitory effects on microglia are therefore considered as a potential strategy to counter balance neurodegenerative progression. Caffeic acid phenethyl ester (CAPE), a natural phenol in honeybee propolis, is known to possess antioxidant, anti-inflammatory and anti-microbial properties. Accordingly, the current study intended to probe the effects of CAPE on microglia activation by using in vitro and in vivo models. Western blot and Griess reaction assay revealed CAPE significantly inhibited the expressions of inducible nitric oxide synthase (NOS), cyclooxygenase (COX)-2 and the production of nitric oxide (NO). Administration of CAPE resulted in increased expressions of hemeoxygenase (HO)-1and erythropoietin (EPO) in microglia. The phosphorylated adenosine monophosphate-activated protein kinase (AMPK)-α was further found to regulate the anti-inflammatory effects of caffeic acid. In vivo results from immunohistochemistry along with rotarod test also revealed the anti-neuroinflammatory effects of CAPE in microglia activation. The current study has evidenced several possible molecular determinants, AMPKα, EPO, and HO-1, in mediating anti-neuroinflammatory responses in microglial cells.


Subject(s)
Caffeic Acids/pharmacology , Microglia/drug effects , Phenylethyl Alcohol/analogs & derivatives , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Erythropoietin/genetics , Erythropoietin/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/metabolism , Male , Mice , Mice, Inbred ICR , Microglia/metabolism , Microglia/pathology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phenylethyl Alcohol/pharmacology
13.
J Neurooncol ; 118(2): 257-269, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24756349

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and lethal type of primary brain tumor characterized by its rapid infiltration to surrounding tissues during the early stages. The fast spreading of GBM obscures the initiation of the tumor mass making the treatment outcome undesirable. Endothelin-1 is known as a secretory protein presented in various types of brain cells, which has been indicated as a factor for cancer pathology. The aim of the present study was to investigate the molecular mechanism of cell migration in GBM. We found that various malignant glioma cells expressed higher amounts of endothelin-1, ETA, and ETB receptors than nonmalignant human astrocytes. The application of endothelin-1 enhanced the migratory activity in human U251 glioma cells corresponding to increased expression of matrix metalloproteinase (MMP)-9 and MMP-13. The endothelin-1-induced cell migration was attenuated by MMP-9 and MMP-13 inhibitors and inhibitors of mitogen-activated protein (MAP) kinase and PI3 kinase/Akt. Furthermore, the elevated levels of phosphate c-Jun accumulation in the nucleus and activator protein-1 (AP-1)-DNA binding activity were also found in endothelin-1 treated glioma cells. In migration-prone sublines, cells with greater migration ability showed higher endothelin-1, ETB receptor, and MMP expressions. These results indicate that endothelin-1 activates MAP kinase and AP-1 signaling, resulting in enhanced MMP-9 and MMP-13 expressions and cell migration in GBM.


Subject(s)
Cell Movement/physiology , Central Nervous System Neoplasms/physiopathology , Endothelin-1/metabolism , Glioblastoma/physiopathology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Astrocytes/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Transcription Factor AP-1/metabolism
14.
Int J Mol Sci ; 15(3): 3889-903, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24599080

ABSTRACT

Glioblastoma multiforme (GBM) is the most common type of primary and malignant tumor occurring in the adult central nervous system. GBM often invades surrounding regions of the brain during its early stages, making successful treatment difficult. Osthole, an active constituent isolated from the dried C. monnieri fruit, has been shown to suppress tumor migration and invasion. However, the effects of osthole in human GBM are largely unknown. Focal adhesion kinase (FAK) is important for the metastasis of cancer cells. Results from this study show that osthole can not only induce cell death but also inhibit phosphorylation of FAK in human GBM cells. Results from this study show that incubating GBM cells with osthole reduces matrix metalloproteinase (MMP)-13 expression and cell motility, as assessed by cell transwell and wound healing assays. This study also provides evidence supporting the potential of osthole in reducing FAK activation, MMP-13 expression, and cell motility in human GBM cells.


Subject(s)
Cell Movement/drug effects , Coumarins/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Matrix Metalloproteinase 13/metabolism , Blotting, Western , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cnidium/chemistry , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Fruit/chemistry , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Phosphorylation/drug effects , Tyrosine/metabolism
15.
Molecules ; 19(7): 8820-39, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24972270

ABSTRACT

Increasing evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play a key role in neurodegeneration. Fisetin, a plant flavonol commonly found in fruits and vegetables, is frequently added to nutritional supplements due to its antioxidant properties. In the present study, treatment with fisetin inhibited microglial cell migration and ROS (reactive oxygen species) production. Treatment with fisetin also effectively inhibited LPS plus IFN-γ-induced nitric oxide (NO) production, and inducible nitric oxide synthase (iNOS) expression in microglial cells. Furthermore, fisetin also reduced expressions of iNOS and NO by stimulation of peptidoglycan, the major component of the Gram-positive bacterium cell wall. Fisetin also inhibited the enhancement of LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator IL (interlukin)-1 ß expression. Besides the antioxidative and anti-inflammatory effects of fisetin, our study also elucidates the manner in fisetin-induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression. Moreover, the regulatory molecular mechanism of fisetin-induced HO-1 expression operates through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Notably, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo. These findings suggest that fisetin may be a candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Microglia/immunology , Neuroprotective Agents/pharmacology , Animals , Cell Line , Cell Movement/drug effects , Drug Evaluation, Preclinical , Flavonols , Heme Oxygenase-1/metabolism , Lipopolysaccharides/pharmacology , Male , Membrane Proteins/metabolism , Mice, Inbred ICR , Microglia/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation
16.
Neurochem Int ; 179: 105811, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39053771

ABSTRACT

Psychological stress induces neuroinflammatory responses, which are associated with the pathogenesis of various psychiatric disorders, such as posttraumatic stress disorder and anxiety. Osthole-a natural coumarin isolated from the seeds of the Chinese herb Cnidium monnieri-exerts anti-inflammatory and antioxidative effects on the central nervous system. However, the therapeutic benefits of osthole against psychiatric disorders remain largely unknown. We previously demonstrated that mice subjected to repeated social defeat stress (RSDS) in the presence of aggressor mice exhibited symptoms of posttraumatic stress disorder, such as social avoidance and anxiety-like behaviors. In this study, we investigated the therapeutic effects of osthole and the underlying molecular mechanisms. Osthole exerted therapeutic effects on cognitive behaviors, mitigating anxiety-like behaviors and social avoidance in a mouse model of RSDS. The anti-inflammatory response induced by the oral administration of osthole was strengthened through the upregulation of heme oxygenase-1 expression. The expression of PPARα was inhibited in mice subjected to RSDS. Nonetheless, osthole treatment reversed the inhibition of PPARα expression. We identified a positive correlation between heme oxygenase-1 expression and PPARα expression in osthole-treated mice. In conclusion, osthole has potential as a Chinese herbal medicine for anxiety disorders. When designing novel drugs for psychiatric disorders, researchers should consider targeting the activation of PPARα.

17.
J Neuroimmune Pharmacol ; 19(1): 38, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066908

ABSTRACT

Repetitive exposure of innate immune cells to a subthreshold dosage of endotoxin components may modulate inflammatory responses. However, the regulatory mechanisms in the interactions between the central nervous system (CNS) and the immune system remain unclear. This study aimed to investigate the effects of lipopolysaccharide (LPS) preconditioning in repeated social defeat stress (RSDS)-induced abnormal immune responses and behavioral impairments. This study aimed to elucidate the mechanisms that underlie the protective effects of repeated administration of a subthreshold dose LPS on behavioral impairments using the RSDS paradigm. LPS preconditioning improved abnormal behaviors in RSDS-defeated mice, accompanied by decreased monoamine oxidases and increased glucocorticoid receptor expression in the hippocampus. In addition, pre-treated with LPS significantly decreased the recruited peripheral myeloid cells (CD11b+CD45hi), mainly circulating inflammatory monocytes (CD11b+CD45hiLy6ChiCCR2+) into the brain in response to RSDS challenge. Importantly, we found that LPS preconditioning exerts its protective properties by regulating lipocalin-2 (LCN2) expression in microglia, which subsequently induces expressions of chemokine CCL2 and pro-inflammatory cytokine. Subsequently, LPS-preconditioning lessened the resident microglia population (CD11b+CD45intCCL2+) in the brains of the RSDS-defeated mice. Moreover, RSDS-associated expressions of leukocytes (CD11b+CD45+CCR2+) and neutrophils (CD11b+CD45+Ly6G+) in the bone marrow, spleen, and blood were also attenuated by LPS-preconditioning. In particular, LPS preconditioning also promoted the expression of endogenous antioxidants and anti-inflammatory proteins in the hippocampus. Our results demonstrate that LPS preconditioning ameliorates lipocalin 2-associated microglial activation and aberrant immune response and promotes the expression of endogenous antioxidants and anti-inflammatory protein, thereby maintaining the homeostasis of pro-inflammation/anti-inflammation in both the brain and immune system, ultimately protecting the mice from RSDS-induced aberrant immune response and behavioral changes.


Subject(s)
Lipopolysaccharides , Mice, Inbred C57BL , Social Defeat , Stress, Psychological , Animals , Lipopolysaccharides/toxicity , Mice , Male , Stress, Psychological/immunology , Microglia/drug effects , Microglia/metabolism , Microglia/immunology , Behavior, Animal/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/immunology , Lipocalin-2/metabolism
18.
Pharmacol Biochem Behav ; 241: 173794, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38834160

ABSTRACT

Psychological stress affects the neuroendocrine regulation, which modulates mental status and behaviors. Melatonin, a hormone synthesized primarily by the pineal gland, regulates many brain functions, including circadian rhythms, pain, sleep, and mood. Selective pharmacological melatonin agonist ramelteon has been clinically used to treat mood and sleep disorders. Posttraumatic stress disorder (PTSD) is a psychiatric condition associated with severe trauma; it is generally triggered by traumatic events, which lead to severe anxiety and uncontrollable trauma recall. We recently reported that repeated social defeat stress (RSDS) may induce robust anxiety-like behaviors and social avoidance in mice. In the present study, we investigated whether melatonin receptor activation by melatonin and ramelteon regulates RSDS-induced behavioral changes. Melatonin treatment improved social avoidance and anxiety-like behaviors in RSDS mice. Moreover, treatment of the non-selective MT1/MT2 receptor agonist, ramelteon, markedly ameliorated RSDS-induced social avoidance and anxiety-like behaviors. Moreover, activating melatonin receptors also balanced the expression of monoamine oxidases, glucocorticoid receptors, and endogenous antioxidants in the hippocampus. Taken together, our findings indicate that the activation of both melatonin and ramelteon regulates RSDS-induced anxiety-like behaviors and PTSD symptoms. The current study also showed that the regulatory effects of neuroendocrine mechanisms and cognitive behaviors on melatonin receptor activation in repeated social defeat stress.


Subject(s)
Anxiety , Indenes , Melatonin , Social Defeat , Stress, Psychological , Animals , Indenes/pharmacology , Mice , Male , Stress, Psychological/metabolism , Stress, Psychological/drug therapy , Melatonin/pharmacology , Anxiety/drug therapy , Anxiety/psychology , Behavior, Animal/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/agonists , Receptor, Melatonin, MT1/agonists , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/agonists , Receptor, Melatonin, MT2/metabolism , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Receptors, Melatonin/agonists , Receptors, Melatonin/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Stress Disorders, Post-Traumatic/metabolism
19.
J Cell Physiol ; 228(3): 563-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22806254

ABSTRACT

Resistin is originally reported as an adipose tissue-specific hormone and is thought to represent a link between obesity and insulin-resistant diabetes. Adipokines exert energy-regulation and has been reported to have neuroprotective effect like leptin, adiponectin, and ghrelin. However, the role of resistin in neuroprotective effect has not been explored. 6-hydroxydopamine (6-OHDA), one of the most investigated Parkinson's disease neurotoxins, is widely used to study mechanisms of cell death in dopaminergic neurons. In the present study, our results show that treatment of resistin protects 6-OHDA-induced cell death in dopaminergic-like MES23.5 cells. Resistin also antagonizes 6-OHDA-induced apoptotic cell death measured by fluorescence-activated cell sorter (FACS) analysis and Hochest 33342 staining. Furthermore, treatment of resistin also dramatically reduces 6-OHDA-mediated ROS production and mitochondria transmembrane potential dissipation. Moreover, expression of 6-OHDA-induced apoptotic markers, such as Bcl-2 degradation, Bax expression, PARP degradation and caspase 3 activity increase, are all attenuated by resistin treatment. Our results also show that resistin induces up-regulation of heat shock protein (Hsp) 32 (heme oxygenase-1, HO-1) and Hsc (heat shock cognate) 70. The protective effect of resistin on 6-OHDA-induced cell death is abolished by HO-1 inhibitor zinc protoporphyrin IX and HSP inhibitor KNK437. These results suggest the neuroprotective effects of resistin against 6-OHDA-induced cell death with the underlying mechanisms of inhibiting oxidative stress and apoptosis. Therefore, we suggest that resistin may provide a useful therapeutic strategy for neurodegenerative diseases such as Parkinson's disease.


Subject(s)
Cell Death/drug effects , Dopaminergic Neurons/drug effects , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Resistin/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Cell Death/physiology , Cell Line , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reactive Oxygen Species/metabolism , Resistin/physiology , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
20.
Toxicol Appl Pharmacol ; 269(1): 43-50, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23500011

ABSTRACT

We investigated the interaction between proinflammatory and inflammatory responses caused by Staphylococcus aureus-derived lipoteichoic acid (LTA) in primary cultured microglial cells and BV-2 microglia. LTA induced inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) protein levels increase in a concentration- and time-dependent manner. Meanwhile, LTA also increased nitric oxide (NO) and PGE2 production in microglia. Administration of TLR2 antagonist effectively inhibited LTA-induced NO, iNOS, and COX-2 expression. Moreover, treatment of cells with LTA caused a time-dependent activation of ERK, p38, JNK, as well as AKT. We also found that LTA-induced iNOS and COX-2 up-regulation were attenuated by p38, JNK, and PI3-kinase inhibitors. On the other hand, LTA-enhanced HO-1 expression was attenuated by p38 and PI3-kinase inhibitors. Treatment of cells with NF-κB and AP-1 inhibitors antagonized LTA-induced iNOS and COX-2 expression. However, only NF-κB inhibitors reduced LTA-induced HO-1 expression in microglia. Furthermore, stimulation of cells with LTA also activated IκBα phosphorylation, p65 phosphorylation at Ser5³6, and c-Jun phosphorylation. Moreover, LTA-induced increases of κB-DNA and AP-1-DNA binding activity were inhibited by p38, JNK, and PI3-kinase inhibitors. HO-1 activator CoPP IX dramatically reversed LTA-induced iNOS expression. Our results provided mechanisms linking LTA and inflammation/anti-inflammation, and indicated that LTA plays a regulatory role in microglia activation.


Subject(s)
Inflammation/immunology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Staphylococcus aureus/metabolism , Teichoic Acids/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Heme Oxygenase (Decyclizing)/metabolism , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Lipopolysaccharides/isolation & purification , Microglia/immunology , Microglia/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Teichoic Acids/isolation & purification , Time Factors , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/metabolism , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL