Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Drug Alcohol Depend ; 257: 111139, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38430788

BACKGROUND: The recreational drug ±3,4-methylenedioxymethamphetamine (MDMA; also known as "ecstasy") has unusual subjective prosocial and empathogenic effects, and has exhibited potential as an adjunct to psychotherapy in recent years. However, there has been some concern regarding possible neuropsychiatric symptoms, such as cognitive impairment and dependence, emerging after abstinence. Therefore, this study aimed to evaluate the mechanism underlying cognitive impairment during MDMA withdrawal. To achieve this, we focused on the arachidonic acid cascade, which is related to addiction to some abusive drugs. METHODS: A novel object recognition task was used to investigate cognitive function in mice. Furthermore, we quantified prostaglandin E2 during MDMA withdrawal. RESULTS: The recognition index significantly decreased during withdrawal after repeated administration of MDMA (10mg/kg, i.p., once daily for 7 days), but not following co-administration of diclofenac (10mg/kg, i.p.), a cyclooxygenase inhibitor. On day 1, following repeated MDMA treatment, prostaglandin E2 content significantly increased in the hippocampus but not in the prefrontal cortex and striatum. CONCLUSIONS: Our findings indicate that activation of the arachidonic acid cascade at least in the hippocampus is likely involved in the development of recognition memory impairment during MDMA withdrawal. Therefore, co-use of cyclooxygenase inhibitors with MDMA may reduce concerns regarding MDMA-induced impairment of recognition memory.


N-Methyl-3,4-methylenedioxyamphetamine , Mice , Animals , N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Arachidonic Acid/pharmacology , Cognition , Hippocampus , Prostaglandins/pharmacology
2.
J Pharmacol Sci ; 153(4): 197-207, 2023 Dec.
Article En | MEDLINE | ID: mdl-37973217

Osteoclasts are multinucleated, specializes bone-resorbing cells that are derived from the monocyte/macrophage lineage. Excessive resorbing activities of osteoclasts are involved in destructive bone diseases. The detailed mechanism of acidification at the bone adhesion surface during the bone resorption process of osteoclasts remains to be defined. During glycolysis, pyruvate proceeds to the tricarboxylic cycle under aerobic conditions and pyruvate is converted to lactate via lactate dehydrogenase A (LDHA) under anaerobic conditions. However, tumor cells produce ATP during aerobic glycolysis and large amounts of pyruvate are converted to lactate and H+ by LDHA. Lactate and H+ are excreted outside the cell, whereby they are involved in invasion of tumor cells due to the pH drop around the cell. In this study, we focused on aerobic glycolysis and investigated the production of lactate by LDHA in osteoclasts. Expression of LDHA and monocarboxylate transporter 4 (MCT4) was upregulated during osteoclast differentiation. Intracellular and extracellular lactate levels increased with upregulation of LDHA and MCT4, respectively. FX11 (an LDHA inhibitor) inhibited osteoclast differentiation and suppressed the bone-resorbing activity of osteoclasts. We propose that inhibition of LDHA may represent a novel therapeutic strategy for controlling excessive bone resorption in osteoporosis and rheumatoid arthritis.


Bone Resorption , Osteogenesis , Humans , Lactate Dehydrogenase 5/metabolism , Osteoclasts/physiology , Bone Resorption/prevention & control , Bone Resorption/metabolism , Lactates/metabolism , Glycolysis , Pyruvates/metabolism , L-Lactate Dehydrogenase/metabolism
3.
Biomed Res ; 41(6): 269-277, 2020.
Article En | MEDLINE | ID: mdl-33268671

Osteoporosis disturbs the balance of bone metabolism, and excessive bone resorption causes a decrease in bone density, thus increasing the risk of fracture. (-)-Epigallocatechin-3-gallate (EGCG) is the most abundant catechin contained in green tea. EGCG has a variety of pharmacological activities. Recently, it was reported that EGCG inhibits osteoclast differentiation, but the details of the mechanism underlying the EGCG-mediated suppression of osteoclastogenesis are unknown. In this study, we investigated the effects of EGCG on several signaling pathways in osteoclastogenesis. EGCG suppressed the expression of the nuclear factor of activated T cells cytoplasmic-1 (NFATc1), the master regulator of osteoclastogenesis. EGCG decreased the expression of cathepsin K, c-Src, and ATP6V0d2 and suppressed bone resorption. We also found that EGCG upregulated heme oxygenase-1 (HO-1) and suppressed the extracellular release of high-mobility group box 1 (HMGB1). In addition, EGCG decreased the expression of the receptor for advanced glycation end products (RAGE), which is the receptor of HMGB1, in osteoclastogenesis. In summary, our study showed that EGCG could inhibit osteoclast differentiation through the downregulation of NFATc1 and the suppression of the HO-1-HMGB1-RAGE pathway. EGCG might have the potential to be a lead compound that suppresses bone resorption in the treatment of osteoporosis.


Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Catechin/analogs & derivatives , Gene Expression Regulation/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoporosis/drug therapy , Animals , Bone Density/drug effects , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , CSK Tyrosine-Protein Kinase/genetics , CSK Tyrosine-Protein Kinase/metabolism , Catechin/pharmacology , Cathepsin K/genetics , Cathepsin K/metabolism , Cell Differentiation/drug effects , Femur/drug effects , Femur/metabolism , Femur/pathology , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Primary Cell Culture , RANK Ligand/antagonists & inhibitors , RANK Ligand/pharmacology , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction , Tibia/drug effects , Tibia/metabolism , Tibia/pathology , Treatment Outcome , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
4.
Biochem Biophys Res Commun ; 530(2): 455-461, 2020 09 17.
Article En | MEDLINE | ID: mdl-32553625

Osteoclasts are multinucleated bone-resorbing cells derived from monocyte/macrophage progenitor cells. Excessive formation and resorbing activities of osteoclasts are involved in the bone-destructive pathologies of rheumatoid arthritis and osteoporosis. Recently, it has been found that nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor for anti-oxidative stress genes, functions in osteoclastogenesis. Dimethyl fumarate (DMF) is a potent activator of Nrf2 and has been shown to inhibit osteoclastogenesis. Here, we investigated the mechanisms of this inhibition by examining the activation of several signalling pathways during the differentiation of bone marrow-derived macrophages into osteoclasts. DMF inhibited the differentiation of osteoclasts in a dose-dependent manner and suppressed the bone-resorbing activity of osteoclasts. DMF treatment decreased the expression of nuclear factor of activated T-cells cytoplasmic-1, and significantly decreased phosphorylation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase in osteoclasts. We also found that DMF inhibited the extracellular release of high mobility group box 1, associated with an up-regulation of heme oxygenase-1, likely mediated through Nrf2 activation. Our results indicate that DMF inhibits osteoclast differentiation through multiple pathways.


Dimethyl Fumarate/pharmacology , HMGB1 Protein/metabolism , MAP Kinase Signaling System/drug effects , NFATC Transcription Factors/metabolism , Osteogenesis/drug effects , Phosphorylation/drug effects , Animals , Cells, Cultured , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HMGB1 Protein/analysis , Male , Mice, Inbred C57BL , NFATC Transcription Factors/analysis , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Am J Chin Med ; 45(7): 1497-1511, 2017.
Article En | MEDLINE | ID: mdl-29025293

Berberine (BBR), an isoquinoline alkaloid, is a well-known bioactive compound contained in medicinal plants used in traditional and folk medicines. In this study, we investigated the subcellular localization and the apoptotic mechanisms of BBR were elucidated. First, we confirmed the incorporation of BBR into the cell visually. BBR showed antiproliferative activity and promptly localized to the nucleus from 5[Formula: see text]min to 15[Formula: see text]min after BBR treatment in HL-60 human promyelocytic leukemia cells. Next, we examined the antiproliferative activity of BBR (1) and its biosynthetically related compounds (2-7) in HL-60 cells. BBR exerted strongest antiproliferative activity among 1-7 and the results of structures and activity relation suggested that a methylenedioxyl group in ring A, an [Formula: see text]-alkyl group at C-9 position, and the frame of isoquinoline may be necessary for antiproliferative activity. Moreover, BBR showed the most potent antiproliferative activity in HL-60 cells among human cancer and normal cell lines tested. Next, we examined the effect of BBR on molecular events known as apoptosis induction. In HL-60 cells, BBR induced chromatin condensation and DNA fragmentation, and triggered the activation of PARP, caspase-3 and caspase-8 without the activation of caspase-9. BBR-induced DNA fragmentation was abolished by pretreatment with inhibitors against caspase-3 and caspase-8, but not against caspase-9. ERK and p38 were promptly phosphorylated after 15 min of BBR treatment, and this was correlated with time of localization to the nucleus of BBR. These results demonstrated that BBR translocated into nucleus immediately after treatments and induced apoptotic cell death by activation of caspase-3 and caspase-8.


Apoptosis/drug effects , Berberine/metabolism , Berberine/pharmacology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Nucleus/metabolism , Leukemia, Promyelocytic, Acute/pathology , Cell Proliferation/drug effects , Cells, Cultured , Chromatin/metabolism , DNA Fragmentation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism , Phosphorylation/drug effects , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Biochem Biophys Res Commun ; 473(2): 620-4, 2016 Apr 29.
Article En | MEDLINE | ID: mdl-27038546

CD147, a membrane glycoprotein of the immunoglobulin superfamily, is highly upregulated during dynamic cellular events including tissue remodelling. Elevated CD147 expression is present in the joint of rheumatoid arthritis patients. However, the role of CD147 in bone destruction remains unclear. To determine whether CD147 is involved in osteoclastogenesis, we studied its expression in mouse osteoclasts and its role in osteoclast differentiation and function. CD147 expression was markedly upregulated during osteoclast differentiation. To investigate the role of CD147 in receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclastogenesis and bone resorption activity, osteoclast precursor cells were transfected with CD147 siRNA. Decreased CD147 expression inhibited osteoclast formation and bone resorption, inhibited RANKL-induced nuclear translocation of the nuclear factor of activated T cells (NFAT) c1 and decreased the expression of the d2 isoform of vacuolar ATPase Vo domain and cathepsin K. Therefore, CD147 plays a critical role in the differentiation and function of osteoclasts by upregulating NFATc1 through the autoamplification of its expression in osteoclastogenesis.


Basigin/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Signal Transduction , Animals , Basigin/genetics , Bone Resorption/genetics , Bone Resorption/metabolism , Cells, Cultured , Male , Mice, Inbred C57BL , Osteoclasts/cytology , Protein Transport , RANK Ligand/metabolism , Up-Regulation
7.
J Inflamm (Lond) ; 10(1): 32, 2013 Oct 09.
Article En | MEDLINE | ID: mdl-24107300

BACKGROUND: BMP4, a member of the transforming growth factor-beta superfamily, is upregulated in the aortas of diabetic db/db mice. However, little is known about its role in diabetic atherosclerosis. Therefore, we examined the roles of BMP4 in the formation of diabetic atherosclerosis in apolipoprotein E knockout (ApoE KO) mice and in the uptake of oxidized low density lipoprotein (oxLDL) in peritoneal macrophages of wild-type mice. METHODS: To induce diabetes, ApoE KO mice were intraperitoneally injected with streptozotocin. Diabetic and non-diabetic ApoE KO mice were then fed a high-fat diet for 4 weeks. Next, to investigate a role of BMP4 in the peritoneal macrophages, we examined the uptake of oxLDL in BMP4-treated macrophages. RESULTS: Diabetic ApoE KO mice showed accelerated progression of aortic plaques accompanied by increased luminal plaque area. Western blot analysis showed that BMP4 expression in the whole aorta was greatly increased in diabetic ApoE KO mice, than non-diabetic mice. Western blot analysis showed that the BMP4/SMAD1/5/8 signaling pathway was strongly activated in the aorta from diabetic ApoE KO mice, compared with control ApoE KO mice. Double immunofluorescence staining showed that BMP4 was expressed in MOMA2-labeled macrophage in the aortic lesions of ApoE KO mice. BMP4 significantly increased the uptake of oxLDL into peritoneal macrophages in vitro. CONCLUSION: We show that in the aorta of diabetic ApoE KO mice, BMP4 is increased and activates SMAD1/5/8. Our in vitro findings indicate that BMP4 enhances oxLDL uptake in mouse peritoneal macrophages, suggesting BMP4 may be involved in aortic plaque formation in diabetic ApoE KO mice. Targeting BMP4 may offer a new strategy for inhibition of plaque progression and stabilization of atherosclerotic lesions.

8.
Biol Pharm Bull ; 36(3): 492-5, 2013.
Article En | MEDLINE | ID: mdl-23449334

The blood-brain barrier (BBB) is formed by brain endothelial cells. Many immortalized brain endothelial cell lines have been established; these have been used as in vitro BBB models. The aim of the present study was to assess the paracellular barrier properties of the immortalized mouse brain endothelial cell lines bEND.3, bEND.5 cells, and mouse brain endothelial cell 4 (MBEC4), and those of the primary mouse brain endothelial cells pMBECs. bEND.3 cells showed low permeability to sodium fluorescein and obvious staining of tight junction proteins (claudin-5, occludin and ZO-1) similar to pMBECs; these barrier properties of MBEC4 and bEND.5 cells were low. In addition, bEND.3 cells expressed the highest level of claudin-5 among all cells. These results suggest that bEND.3 cells are a convenient and useful model for evaluating BBB function, especially the paracellular barrier.


Blood-Brain Barrier , Brain/blood supply , Endothelial Cells/metabolism , Tight Junction Proteins/analysis , Animals , Cell Line , Cell Survival , Claudin-5/analysis , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Occludin/analysis , Zonula Occludens-1 Protein/analysis
9.
J Inflamm (Lond) ; 9(1): 44, 2012 Nov 20.
Article En | MEDLINE | ID: mdl-23167819

BACKGROUND: Cyclophilin A (CypA), a member of the immunophilin family, is a ubiquitously distributed intracellular protein. Recent studies have shown that CypA is secreted by cells in response to inflammatory stimuli. Elevated levels of extracellular CypA and its receptor, CD147 have been detected in the synovium of patients with RA. However, the precise process of interaction between CypA and CD147 in the development of RA remains unclear. This study aimed to investigate CypA secretion from fibroblast-like synoviocytes (FLS) isolated from mice with collagen-induced arthritis (CIA) and CypA-induced CD147 expression in mouse macrophages. FINDINGS: CIA was induced by immunization with type II collagen in mice. The expression and localization of CypA and CD147 was investigated by immunoblotting and immunostaining. Both CypA and CD147 were highly expressed in the joints of CIA mice. CD147 was expressed in the infiltrated macrophages in the synovium of CIA mice. In vitro, spontaneous CypA secretion from FLS was detected and this secretion was increased by stimulation with lipopolysaccharide. CypA markedly increased CD147 levels in macrophages. CONCLUSIONS: These findings suggest that an interaction in the synovial joints between extracellular CypA and CD147 expressed by macrophages may be involved in the mechanisms underlying the development of arthritis.

10.
Neurosci Lett ; 531(2): 114-9, 2012 Dec 07.
Article En | MEDLINE | ID: mdl-23127852

Recently, 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase inhibitors were reported to induce neurite outgrowth in vitro. However, the mechanism underlying this effect remains unclear. Cellular prion protein (PrP(C)) is a ubiquitous glycoprotein present on the surfaces of various cells, including neurons, and is suggested to be involved in neurite outgrowth. Therefore, the present study aimed to determine whether PrP(C) mediates neurite outgrowth induced by HMG-CoA reductase inhibitors. Atorvastatin, a strong HMG-CoA reductase inhibitor, induced neurite outgrowth and increased PrP(C) levels in Neuro2a cells in a time- and dose-dependent manner. PrP(C) mRNA expression was also increased by atorvastatin. Farnesol, a non-sterol mevalonate derivative, attenuated the atorvastatin-induced neurite outgrowth and increase in PrP(C). Neuro2a cells overexpressing PrP(C) showed a remarkable enhancement of atorvastatin-induced neurite outgrowth compared with mock cells transfected with empty pCI-neo vector. These findings suggest that PrP(C) contributes, at least in part, to atorvastatin-induced neurite outgrowth. This phenomenon may be included among the mechanisms underlying decreased risk of Alzheimer's disease in patients treated with HMG-CoA reductase inhibitors.


Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurites/metabolism , Prions/metabolism , Pyrroles/pharmacology , Animals , Atorvastatin , Cell Line, Tumor , Mice , Neurites/drug effects , Neuroblastoma , Reverse Transcriptase Polymerase Chain Reaction
11.
Neurosci Lett ; 524(1): 45-8, 2012 Aug 22.
Article En | MEDLINE | ID: mdl-22801252

P-glycoprotein, an efflux transporter that is highly expressed at the blood-brain barrier (BBB), is involved in the traffic of several compounds across the BBB. BBB disruption under pathological conditions is observed in parallel with microglial activation. Previous studies of the interaction between rat brain endothelial cells (RBECs) and microglia have shown that lipopolysaccharide (LPS) activated microglia increase the permeability of RBECs through a mechanism involving NADPH oxidase. In this study, to investigate whether LPS-activated microglia are linked to P-gp dysfunction at the BBB, we examined the effect of LPS on P-gp function in a coculture system with RBECs and rat microglia. When LPS at a concentration showing no effect on the RBEC monolayer was added for 6h to the abluminal side of the RBEC monolayer and RBEC/microglia cocultures, cellular accumulation of the P-gp substrate rhodamine 123, in RBECs, was increased by LPS in the RBEC/microglia coculture. This increased accumulation of rhodamine 123 in RBECs was blocked by diphenyleneiodoniumchloride, an NADPH oxidase inhibitor. P-gp expression on RBECs was not influenced by treatment with LPS in either RBEC monolayers or RBEC/microglia cocultures. These findings suggest that activated microglia induce P-gp dysfunction at the BBB through an NADPH oxidase-dependent pathway.


ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/blood supply , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism , Microvessels/metabolism , Animals , Animals, Newborn , Blood-Brain Barrier/metabolism , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Fluorescent Dyes , Microglia/drug effects , Microvessels/drug effects , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Rats , Rats, Wistar , Rhodamine 123
12.
J Neuroinflammation ; 8: 106, 2011 Aug 26.
Article En | MEDLINE | ID: mdl-21867555

BACKGROUND: Increased matrix metalloproteinase (MMP)-9 in the plasma and brain is associated with blood-brain barrier (BBB) disruption through proteolytic activity in neuroinflammatory diseases. MMP-9 is present in the brain microvasculature and its vicinity, where brain microvascular endothelial cells (BMECs), pericytes and astrocytes constitute the BBB. Little is known about the cellular source and role of MMP-9 at the BBB. Here, we examined the ability of pericytes to release MMP-9 and migrate in response to inflammatory mediators in comparison with BMECs and astrocytes, using primary cultures isolated from rat brains. METHODS: The culture supernatants were collected from primary cultures of rat brain endothelial cells, pericytes, or astrocytes. MMP-9 activities and levels in the supernatants were measured by gelatin zymography and western blot, respectively. The involvement of signaling molecules including mitogen-activated protein kinases (MAPKs) and phosphoinositide-3-kinase (PI3K)/Akt in the mediation of tumor necrosis factor (TNF)-α-induced MMP-9 release was examined using specific inhibitors. The functional activity of MMP-9 was evaluated by a cell migration assay. RESULTS: Zymographic and western blot analyses demonstrated that TNF-α stimulated pericytes to release MMP-9, and this release was much higher than from BMECs or astrocytes. Other inflammatory mediators [interleukin (IL)-1ß, interferon-γ, IL-6 and lipopolysaccharide] failed to induce MMP-9 release from pericytes. TNF-α-induced MMP-9 release from pericytes was found to be mediated by MAPKs and PI3K. Scratch wound healing assay showed that in contrast to BMECs and astrocytes the extent of pericyte migration was significantly increased by TNF-α. This pericyte migration was inhibited by anti-MMP-9 antibody. CONCLUSION: These findings suggest that pericytes are most sensitive to TNF-α in terms of MMP-9 release, and are the major source of MMP-9 at the BBB. This pericyte-derived MMP-9 initiated cellular migration of pericytes, which might be involved in pericyte loss in the damaged BBB.


Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cell Movement/physiology , Matrix Metalloproteinase 9/metabolism , Pericytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Culture Media/chemistry , Enzyme Inhibitors/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Pericytes/cytology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism
13.
Cell Mol Neurobiol ; 31(7): 985-9, 2011 Oct.
Article En | MEDLINE | ID: mdl-21681559

Olanzapine is known to be advantageous with respect to outcome and drug compliance in patients with schizophrenia. However, olanzapine has adverse effects, including a higher incidence of weight gain and metabolic disturbances, when compared with those of other antipsychotic agents. The mechanisms underlying these adverse events remain obscure. Female rats were orally administered olanzapine (2 mg/kg) or vehicle once a day for 2 weeks to ascertain if hypothalamic AMP-activated protein kinase (AMPK) mediates olanzapine-induced weight gain and hyperphagia. Body weight and food intake in each rat were evaluated every day and every two days, respectively. After the termination of drug treatment, we measured the protein levels of AMPK and phosphorylated AMPK in the hypothalamus using western blot analyses. Olanzapine significantly increased body weight and food intake. The phosphorylation levels of AMPK were significantly elevated by olanzapine. These results suggest that activation of hypothalamic AMPK may mediate hyperphagia and weight gain induced by chronic treatment with olanzapine.


AMP-Activated Protein Kinases/metabolism , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Hyperphagia/chemically induced , Hypothalamus/drug effects , Hypothalamus/enzymology , Weight Gain/drug effects , Animals , Body Weight/drug effects , Eating/drug effects , Female , Hyperphagia/metabolism , Olanzapine , Rats , Rats, Sprague-Dawley
14.
Neurosci Lett ; 496(2): 121-4, 2011 Jun 01.
Article En | MEDLINE | ID: mdl-21511010

The conversion of cellular prion protein (PrP(C)) to its protease-resistant isoform is involved in the pathogenesis of prion disease. Although PrP(C) is a ubiquitous glycoprotein that is present in various cell types, the physiological role of PrP(C) remains obscure. The present study aimed to determine whether PrP(C) mediates migration of brain microvascular endothelial cells. Small interfering RNAs (siRNAs) targeting PrP(C) were transfected into a mouse brain microvascular endothelial cell line (bEND.3 cells). siPrP1, selected among three siRNAs, reduced mRNA and protein levels of PrP(C) in bEND.3 cells. Cellular migration was evaluated with a scratch-wound assay. siPrP1 suppressed migration without significantly affecting cellular proliferation. This study provides the first evidence that PrP(C) may be necessary for brain microvascular endothelial cells to migrate into damaged regions in the brain. This function of PrP(C) in the brain endothelium may be a mechanism by which the neurovascular unit recovers from an injury such as an ischemic insult.


Brain/physiology , Cell Movement/physiology , Endothelial Cells/physiology , Microvessels/physiology , PrPC Proteins/metabolism , Animals , Cell Line , Cell Proliferation , Mice , Organ Specificity , Tissue Distribution
15.
Life Sci ; 88(11-12): 529-34, 2011 Mar 14.
Article En | MEDLINE | ID: mdl-21262237

AIMS: Cyclosporin A, a calcineurin inhibitor, produces neurotoxicity with relatively high frequency in organ-transplanted patients. The aim of the present study was to clarify whether acute liver failure (ALF) simulated to the transient liver dysfunction at an early phase after liver transplantation increases the susceptibility to cyclosporin A-induced neurotoxicity through the blood-brain barrier (BBB) dysfunction. MAIN METHODS: The right internal, left lateral and left internal lobes in male ddy mice were surgically excised under sodium pentobarbital anesthesia. Effect of cyclosporin A on harmine-induced tremors was examined and BBB permeability to (3)[H]cyclosporin A was assessed in partially (70%) hepatectomized mice at postoperative days 1, 3 and 7. KEY FINDINGS: Patrial hepatectomy aggravated harmine-induced tremors. Cyclosporin A (50mg/kg, i.p.) markedly augmented harmine-induced tremors in partially hepatectomized mice at postoperative day 1. Consistent with these behavioral findings, the brain uptake of (3)[H]cyclosporin A in mice injected with (3)[H]cyclosporin A into the jugular vein at postoperative day 1 was significantly increased by partial hepatectomy compared with sham operation. SIGNIFICANCE: Our results indicate that ALF increases BBB permeability to cyclosporin A by lowering the function of P-glycoprotein and tight junctions, consequently leading to augmentation of cyclosporin A-induced neurotoxicity. The possibility that cyclosporin A increases the risk of neurotoxicity including tremors at an early phase of liver transplantation must be considered.


Blood-Brain Barrier/drug effects , Cyclosporine/adverse effects , Hepatectomy/adverse effects , Immunosuppressive Agents/adverse effects , Neurotoxicity Syndromes/etiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Behavior, Animal/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiology , Capillary Permeability/drug effects , Cell Culture Techniques , Cell Line , Cyclosporine/pharmacokinetics , Harmine/pharmacology , Immunosuppressive Agents/pharmacokinetics , Liver Failure, Acute/blood , Liver Failure, Acute/etiology , Liver Failure, Acute/metabolism , Liver Function Tests , Liver Transplantation/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Neurotoxicity Syndromes/blood , Neurotoxicity Syndromes/metabolism , Tremor/chemically induced
16.
Microvasc Res ; 81(1): 103-7, 2011 Jan.
Article En | MEDLINE | ID: mdl-21036181

The blood-brain barrier (BBB) is the interface that separates the central nervous system (CNS) from the peripheral circulation. An increase in blood-borne substances including cytokines in plasma and brain affects BBB function, and this is associated with the development of pathogenesis of a number of diseases. Plasminogen activator inhibitor (PAI)-1 regulates the plasminogen activator/plasmin system as a serpin in the periphery and the CNS. We investigated whether PAI-1 alters BBB function using in vitro models of the BBB consisting of rat primary brain endothelial cells (RBECs) alone and co-cultured with pericytes. We found that PAI-1 increased the tightness of the brain endothelial barrier in a time- and dose-dependent manner, as shown by an increase in the transendothelial electrical resistance (TEER) and a decrease in the permeability to sodium fluorescein (Na-F). RBECs responded equally to PAI-1 in the blood-facing and brain-facing sides of the brain, leading to a decrease in Na-F permeability. In addition, RBECs constitutively released PAI-1 into the blood-facing (luminal) and brain-facing (abluminal) sides. This release was polarized in favor of the luminal side and facilitated by serum. The neutralization of PAI-1 by an antibody to PAI-1 in RBEC/pericyte co-culture more robustly reduced TEER of RBECs than in RBEC monolayers. These findings suggest that PAI-1 derived from the neurovascular unit and peripheral vascular system participates as a positive regulator of the BBB in facilitating the barrier function of the endothelial tight junctions.


Autocrine Communication/physiology , Blood-Brain Barrier/physiology , Paracrine Communication/physiology , Plasminogen Activator Inhibitor 1/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Blood-Brain Barrier/drug effects , Brain/cytology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free/metabolism , Culture Media, Serum-Free/pharmacology , Electric Impedance , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluorescein/metabolism , Pericytes/cytology , Pericytes/drug effects , Pericytes/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/immunology , Plasminogen Activator Inhibitor 1/pharmacology , Rats , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Serum/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Neurosci Lett ; 482(3): 208-11, 2010 Oct 04.
Article En | MEDLINE | ID: mdl-20654699

Patients with rheumatoid arthritis (RA) are at higher risk of developing pathological cardiovascular and cerebrovascular events than non-RA subjects. Vascular endothelial dysfunction is involved in the induction of cardiovascular events and this process is also observed in patients with RA. Endothelial dysfunction impairs the integrity of the blood-brain barrier (BBB); this phenomenon also underlies brain damage in cerebrovascular diseases. This study was aimed at evaluating the influence of a chronic inflammatory state on BBB integrity in RA using collagen-induced arthritis (CIA), an animal model of RA. CIA was induced by intradermal injection of type II collagen emulsified with Freund's complete adjuvant at the base of the tail of DBA/1 mice. Cerebrovascular permeability was assessed by measurement of sodium fluorescein (Na-F) content in the brains of CIA mice. The expression level of tight junction proteins was investigated by immunoblotting and immunofluorescence of occludin and zonula occludens-1 (ZO-1). Cerebrovascular permeability to Na-F in the brain was increased in CIA mice. This CIA-induced BBB hyperpermeability was more remarkable in the advanced stage than that in the persistent stage of the arthritis. The expression of occludin, but not that of ZO-1, was decreased by CIA. Our results indicate that the integrity of the BBB could be impaired in the inflammatory pathophysiology of RA.


Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Blood-Brain Barrier/pathology , Tight Junctions/pathology , Animals , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Blood-Brain Barrier/metabolism , Fluorescent Antibody Technique , Immunoblotting , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred DBA , Occludin , Phosphoproteins/biosynthesis , Tight Junctions/metabolism , Zonula Occludens-1 Protein
18.
Eur J Pharmacol ; 644(1-3): 5-9, 2010 Oct 10.
Article En | MEDLINE | ID: mdl-20553921

Cyclosporin A, a potent immunosuppressant, can often produce neurotoxicity in patients, although its penetration into the brain is restricted by the blood-brain barrier (BBB). Brain pericytes and astrocytes, which are periendothelial accessory structures of the BBB, can be involved in cyclosporin A-induced BBB disruption. However, the mechanism by which cyclosporin A causes BBB dysfunction remains unknown. Here, we show that in rodent brain endothelial cells, cyclosporin A decreased transendothelial electrical resistance (TEER) by inhibiting intracellular signal transduction downstream of adrenomedullin, an autocrine regulator of BBB function. Cyclosporin A stimulated adrenomedullin release from brain endothelial cells, but did not affect binding of adrenomedullin to its receptors. This cyclosporin A-induced decrease in TEER was attenuated by exogenous addition of adrenomedullin. Cyclosporin A dose-dependently decreased the total cAMP concentration in brain endothelial cells. A combination of cyclosporin A (1microM) with an adenylyl cyclase inhibitor, 9-(tetrahydro-2-furanyl)-9H-purin-6-amine (SQ22536; 10microM), or a protein kinase A (PKA) inhibitor, N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide dihydrochloride (H89; 1microM), markedly increased sodium fluorescein permeability in brain endothelial cells, whereas each drug alone had no effect. Thus, these data suggest that cyclosporin A inhibits the adenylyl cyclase/cyclic AMP/PKA signaling pathway activated by adrenomedullin, leading to impairment of brain endothelial barrier function.


Blood-Brain Barrier/drug effects , Cyclosporine/toxicity , Endothelial Cells/drug effects , Immunosuppressive Agents/toxicity , Adenylyl Cyclases/drug effects , Adenylyl Cyclases/metabolism , Adrenomedullin/administration & dosage , Adrenomedullin/metabolism , Animals , Autocrine Communication , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclosporine/administration & dosage , Cyclosporine/pharmacokinetics , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/metabolism , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Mice , Mice, Inbred BALB C , Permeability , Up-Regulation/drug effects
19.
J Pharmacol Sci ; 112(2): 251-4, 2010.
Article En | MEDLINE | ID: mdl-20118615

The present study was designed to elucidate the involvement of tumor necrosis factor-alpha (TNF-alpha) release from activated microglia in the induction of blood-brain barrier (BBB) dysfunction in an in vitro co-culture system with mouse brain capillary endothelial cells (MBEC4) and microglia. Lipopolysaccharide (LPS)-activated microglia increased the permeability of MBEC4 cells to sodium-fluorescein, and this hyper-permeability was blocked by a neutralizing antibody against TNF-alpha. LPS stimulated microglia to facilitate TNF-alpha release. These findings suggested that TNF-alpha released from activated microglia is attributable to BBB dysfunction.


Blood-Brain Barrier/physiopathology , Microglia/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/metabolism , Capillaries/metabolism , Cell Membrane Permeability , Coculture Techniques , Endothelium, Vascular/metabolism , Fluorescein/pharmacokinetics , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C
20.
Cell Mol Neurobiol ; 30(2): 247-53, 2010 Mar.
Article En | MEDLINE | ID: mdl-19728078

The blood-brain barrier (BBB) is formed by brain capillary endothelial cells, astrocytes, pericytes, microglia, and neurons. BBB disruption under pathological conditions such as neurodegenerative disease and inflammation is observed in parallel with microglial activation. To test whether activation of microglia is linked to BBB dysfunction, we evaluated the effect of lipopolysaccharide (LPS) on BBB functions in an in vitro co-culture system with rat brain microvascular endothelial cells (RBEC) and microglia. When LPS was added for 6 h to the abluminal side of RBEC/microglia co-culture at a concentration showing no effects on the RBEC monolayer, transendothelial electrical resistance was decreased and permeability to sodium-fluorescein was increased in RBEC. Immunofluorescence staining for tight junction proteins demonstrated that zonula occludens-1-, claudin-5-, and occludin-like immunoreactivities at the intercellular borders of RBEC were fragmented in the presence of LPS-activated microglia. These functional changes induced by LPS-activated microglia were blocked by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor, diphenyleneiodonium chloride. The present findings suggest that LPS activates microglia to induce dysfunction of the BBB by producing reactive oxygen species through NADPH oxidase.


Blood-Brain Barrier , Coculture Techniques , Endothelial Cells/physiology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/physiology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Cell Membrane Permeability , Endothelial Cells/cytology , Microglia/cytology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
...