Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
Glia ; 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39166289

RÉSUMÉ

Na+-K+-2Cl- cotransporter-1 (NKCC1) is present in brain cells, including astrocytes. The expression of astrocytic NKCC1 increases in the acute phase of traumatic brain injury (TBI), which induces brain edema. Endothelin-1 (ET-1) is a factor that induces brain edema and regulates the expression of several pathology-related genes in astrocytes. In the present study, we investigated the effect of ET-1 on NKCC1 expression in astrocytes. ET-1 (100 nM)-treated cultured astrocytes showed increased NKCC1 mRNA and protein levels. The effect of ET-1 on NKCC1 expression in cultured astrocytes was reduced by BQ788 (1 µM), an ETB antagonist, but not by FR139317 (1 µM), an ETA antagonist. The involvement of ET-1 in NKCC1 expression in TBI was examined using a fluid percussion injury (FPI) mouse model that replicates the pathology of TBI with high reproducibility. Administration of BQ788 (15 nmol/day) decreased FPI-induced expressions of NKCC1 mRNA and protein, accompanied with a reduction of astrocytic activation. FPI-induced brain edema was attenuated by BQ788 and NKCC1 inhibitors (azosemide and bumetanide). ET-1-treated cultured astrocytes showed increased mRNA and protein expression of hypoxia-inducible factor-1α (HIF1α). Immunohistochemical observations of mouse cerebrum after FPI showed co-localization of HIF1α with GFAP-positive astrocytes. Increased HIF1α expression in the TBI model was reversed by BQ788. FM19G11 (an HIF inhibitor, 1 µM) and HIF1α siRNA suppressed ET-induced increase in NKCC1 expression in cultured astrocytes. These results indicate that ET-1 increases NKCC1 expression in astrocytes through the activation of HIF1α.

2.
Vitam Horm ; 126: 97-111, 2024.
Article de Anglais | MEDLINE | ID: mdl-39029978

RÉSUMÉ

Sonic hedgehog (Shh) is a secreted glycopeptide belonging to the hedgehog family that is essential for morphogenesis during embryonic development. The Shh signal is mediated by two membrane proteins, Patched-1 (Ptch-1) and Smoothened (Smo), following the activation of transcription factors such as Gli. Shh decreases the permeability of the blood-brain barrier (BBB) and plays a key role in its function. In the damaged brain, BBB function is remarkably disrupted. The BBB disruption causes brain edema and neuroinflammation resulting from the extravasation of serum components and the infiltration of inflammatory cells into the cerebral parenchyma. Multiple studies have suggested that astrocyte is a source of Shh and that astrocytic Shh production is increased in the damaged brain. In various experimental animal models of acute brain injury, Shh or Shh signal activators alleviate BBB disruption by increasing tight junction proteins in endothelial cells. Furthermore, activation of astrocytic Shh signaling reduces reactive astrogliosis, neuroinflammation, and increases the production of vascular protective factors, which alleviates BBB disruption in the damaged brain. These findings suggest that astrocytic Shh and Shh signaling protect BBB function in the damaged brain and that target drugs for Shh signaling are expected to be novel therapeutic drugs for acute brain injuries.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Protéines Hedgehog , Transduction du signal , Protéines Hedgehog/métabolisme , Barrière hémato-encéphalique/métabolisme , Astrocytes/métabolisme , Animaux , Humains , Transduction du signal/physiologie
3.
Int J Mol Sci ; 25(6)2024 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-38542369

RÉSUMÉ

Arrestins are known to be involved not only in the desensitization and internalization of G protein-coupled receptors but also in the G protein-independent activation of mitogen-activated protein (MAP) kinases, such as extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), to regulate cell proliferation and inflammation. Our previous study revealed that the histamine H1 receptor-mediated activation of ERK is dually regulated by Gq proteins and arrestins. In this study, we investigated the roles of Gq proteins and arrestins in the H1 receptor-mediated activation of JNK in Chinese hamster ovary (CHO) cells expressing wild-type (WT) human H1 receptors, the Gq protein-biased mutant S487TR, and the arrestin-biased mutant S487A. In these mutants, the Ser487 residue in the C-terminus region of the WT was truncated (S487TR) or mutated to alanine (S487A). Histamine significantly stimulated JNK phosphorylation in CHO cells expressing WT and S487TR but not S487A. Histamine-induced JNK phosphorylation in CHO cells expressing WT and S487TR was suppressed by inhibitors against H1 receptors (ketotifen and diphenhydramine), Gq proteins (YM-254890), and protein kinase C (PKC) (GF109203X) as well as an intracellular Ca2+ chelator (BAPTA-AM) but not by inhibitors against G protein-coupled receptor kinases (GRK2/3) (cmpd101), ß-arrestin2 (ß-arrestin2 siRNA), and clathrin (hypertonic sucrose). These results suggest that the H1 receptor-mediated phosphorylation of JNK is regulated by Gq-protein/Ca2+/PKC-dependent but GRK/arrestin/clathrin-independent pathways.


Sujet(s)
Arrestine , Histamine , Animaux , Cricetinae , Humains , Arrestine/métabolisme , Arrestines/métabolisme , bêta-Arrestines/métabolisme , Cellules CHO , Clathrine/métabolisme , Cricetulus , Extracellular Signal-Regulated MAP Kinases/métabolisme , Kinases associées à des récepteurs couplés à une protéine G/métabolisme , Protéines G/métabolisme , Histamine/pharmacologie , Histamine/métabolisme , Phosphorylation , Protéine kinase C/métabolisme , Récepteur histaminergique H1/génétique , Récepteur histaminergique H1/métabolisme , Transduction du signal
4.
FASEB J ; 38(2): e23425, 2024 01 31.
Article de Anglais | MEDLINE | ID: mdl-38226852

RÉSUMÉ

Postprandial hyperglycemia is an early indicator of impaired glucose tolerance that leads to type 2 diabetes mellitus (T2DM). Alterations in the fatty acid composition of phospholipids have been implicated in diseases such as T2DM and nonalcoholic fatty liver disease. Lysophospholipid acyltransferase 10 (LPLAT10, also called LPCAT4 and LPEAT2) plays a role in remodeling fatty acyl chains of phospholipids; however, its relationship with metabolic diseases has not been fully elucidated. LPLAT10 expression is low in the liver, the main organ that regulates metabolism, under normal conditions. Here, we investigated whether overexpression of LPLAT10 in the liver leads to improved glucose metabolism. For overexpression, we generated an LPLAT10-expressing adenovirus (Ad) vector (Ad-LPLAT10) using an improved Ad vector. Postprandial hyperglycemia was suppressed by the induction of glucose-stimulated insulin secretion in Ad-LPLAT10-treated mice compared with that in control Ad vector-treated mice. Hepatic and serum levels of phosphatidylcholine 40:7, containing C18:1 and C22:6, were increased in Ad-LPLAT10-treated mice. Serum from Ad-LPLAT10-treated mice showed increased glucose-stimulated insulin secretion in mouse insulinoma MIN6 cells. These results indicate that changes in hepatic phosphatidylcholine species due to liver-specific LPLAT10 overexpression affect the pancreas and increase glucose-stimulated insulin secretion. Our findings highlight LPLAT10 as a potential novel therapeutic target for T2DM.


Sujet(s)
1-Acylglycerophosphocholine acyltransferase , Diabète de type 2 , Intolérance au glucose , Animaux , Souris , 1-Acylglycerophosphocholine acyltransferase/génétique , Glucose/pharmacologie , Sécrétion d'insuline , Foie , Phosphatidylcholines , Phospholipides
5.
Biol Pharm Bull ; 47(2): 350-360, 2024.
Article de Anglais | MEDLINE | ID: mdl-38296549

RÉSUMÉ

Traumatic brain injury (TBI) is severe damage to the head caused by traffic accidents, falls, and sports. Because TBI-induced disruption of the blood-brain barrier (BBB) causes brain edema and neuroinflammation, which are major causes of death or serious disabilities, protection and recovery of BBB function may be beneficial therapeutic strategies for TBI. Astrocytes are key components of BBB integrity, and astrocyte-derived bioactive factors promote and suppress BBB disruption in TBI. Therefore, the regulation of astrocyte function is essential for BBB protection. In the injured cerebrum of TBI model mice, we found that the endothelin ETB receptor, histamine H2 receptor, and transient receptor potential vanilloid 4 (TRPV4) were predominantly expressed in reactive astrocytes. We also showed that repeated administration of an ETB receptor antagonist, H2 receptor agonist, and TRPV4 antagonist alleviated BBB disruption and brain edema in a TBI mouse model. Furthermore, these drugs decreased the expression levels of astrocyte-derived factors promoting BBB disruption and increased the expression levels of astrocyte-derived protective factors in the injured cerebrum after TBI. These results suggest that the ETB receptor, H2 receptor, and TRPV4 are molecules that regulate astrocyte function, and might be attractive candidates for the development of therapeutic drugs for TBI.


Sujet(s)
Oedème cérébral , Lésions traumatiques de l'encéphale , Souris , Animaux , Astrocytes/métabolisme , Oedème cérébral/étiologie , Canaux cationiques TRPV/métabolisme , Lésions traumatiques de l'encéphale/traitement médicamenteux , Barrière hémato-encéphalique/métabolisme
6.
Biochem Pharmacol ; 213: 115595, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37201878

RÉSUMÉ

Gq protein-coupled histamine H1 receptors play crucial roles in allergic and inflammatory reactions, in which the phosphorylation of extracellular signal-regulated kinase (ERK) appears to mediate the production of inflammatory cytokines. ERK phosphorylation is regulated by G protein- and arrestin-mediated signal transduction pathways. Here, we aimed to explore how H1 receptor-mediated processes of ERK phosphorylation might be differentially regulated by Gq proteins and arrestins. For this purpose, we evaluated the regulatory mechanism(s) of H1 receptor-mediated ERK phosphorylation in Chinese hamster ovary cells expressing Gq protein- and arrestin-biased mutants of human H1 receptors, S487TR and S487A, in which the Ser487 residue in the C-terminal was truncated and mutated to alanine, respectively. Immunoblotting analysis indicated that histamine-induced ERK phosphorylation was prompt and transient in cells expressing Gq protein-biased S487TR, whereas it was slow and sustained in cells expressing arrestin-biased S487A. Inhibitors of Gq proteins (YM-254890) and protein kinase C (PKC) (GF109203X), and an intracellular Ca2+ chelator (BAPTA-AM) suppressed histamine-induced ERK phosphorylation in cells expressing S487TR, but not those expressing S487A. Conversely, inhibitors of G protein-coupled receptor kinases (GRK2/3) (cmpd101), ß-arrestin2 (ß-arrestin2 siRNA), clathrin (hypertonic sucrose), Raf (LY3009120), and MEK (U0126) suppressed histamine-induced ERK phosphorylation in cells expressing S487A, but not those expressing S487TR. These results suggest that H1 receptor-mediated ERK phosphorylation might be differentially regulated by the Gq protein/Ca2+/PKC and GRK/arrestin/clathrin/Raf/MEK pathways to potentially determine the early and late phases of histamine-induced allergic and inflammatory responses, respectively.


Sujet(s)
Arrestines , Extracellular Signal-Regulated MAP Kinases , Animaux , Cricetinae , Humains , Arrestine/métabolisme , Arrestines/génétique , Arrestines/métabolisme , Cellules CHO , Clathrine/métabolisme , Cricetulus , Extracellular Signal-Regulated MAP Kinases/métabolisme , Protéines G/métabolisme , Histamine/pharmacologie , Histamine/métabolisme , Mitogen-Activated Protein Kinase Kinases/métabolisme , Phosphorylation , Protéine kinase C/métabolisme , Récepteurs couplés aux protéines G/métabolisme
7.
Cells ; 12(5)2023 02 24.
Article de Anglais | MEDLINE | ID: mdl-36899860

RÉSUMÉ

Traumatic brain injury (TBI) is an intracranial injury caused by accidents, falls, or sports. The production of endothelins (ETs) is increased in the injured brain. ET receptors are classified into distinct types, including ETA receptor (ETA-R) and ETB receptor (ETB-R). ETB-R is highly expressed in reactive astrocytes and upregulated by TBI. Activation of astrocytic ETB-R promotes conversion to reactive astrocytes and the production of astrocyte-derived bioactive factors, including vascular permeability regulators and cytokines, which cause blood-brain barrier (BBB) disruption, brain edema, and neuroinflammation in the acute phase of TBI. ETB-R antagonists alleviate BBB disruption and brain edema in animal models of TBI. The activation of astrocytic ETB receptors also enhances the production of various neurotrophic factors. These astrocyte-derived neurotrophic factors promote the repair of the damaged nervous system in the recovery phase of patients with TBI. Thus, astrocytic ETB-R is expected to be a promising drug target for TBI in both the acute and recovery phases. This article reviews recent observations on the role of astrocytic ETB receptors in TBI.


Sujet(s)
Oedème cérébral , Lésions traumatiques de l'encéphale , Animaux , Astrocytes/métabolisme , Encéphale/métabolisme , Oedème cérébral/étiologie , Lésions traumatiques de l'encéphale/complications , Endothélines/métabolisme , Humains
8.
J Pharmacol Sci ; 150(3): 135-145, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36184118

RÉSUMÉ

Histamine is a major neurotransmitter and alleviates neuronal damage after ischemic injury via H2 receptors. Herein, we investigated the effects of H2 receptor agonists on the blood-brain barrier (BBB) disruption after traumatic brain injury (TBI). Male ddY mice were used to generate the TBI model, in which a fluid percussion injury (FPI) was induced by a hydraulic impact. The BBB disruption was evaluated using Evans blue extravasation. H2 receptor agonists, amthamine and dimaprit, were administered into the lateral cerebroventricle (i.c.v.) or tail vein (i.v.) from 3 hours to 3 days after FPI. The i.c.v. or i.v. administration of amthamine and dimaprit reduced FPI-induced Evans blue extravasation and promoted mRNA expression of vascular protective factors, including angiopoietin-1 and sonic hedgehog. The co-administration of ranitidine, a H2 receptor antagonist, inhibited these effects. Expression of the H2 receptor was observed in astrocytes and brain microvascular endothelial cells (BMECs) in the injured cortex. Treatment with amthamine and dimaprit promoted mRNA expression of vascular protective factors in astrocytes and BMECs. These results suggest that H2 receptor agonists alleviate TBI-induced BBB disruption by increasing the expression of vascular protective factors in astrocytes and BMECs.


Sujet(s)
Lésions traumatiques de l'encéphale , Agonistes histaminergiques , Angiopoïétine-1/métabolisme , Angiopoïétine-1/pharmacologie , Animaux , Barrière hémato-encéphalique/métabolisme , Lésions traumatiques de l'encéphale/traitement médicamenteux , Lésions traumatiques de l'encéphale/métabolisme , Dimaprit/métabolisme , Dimaprit/pharmacologie , Cellules endothéliales/métabolisme , Bleu d'Evans/métabolisme , Bleu d'Evans/pharmacologie , Protéines Hedgehog , Histamine/pharmacologie , Agonistes histaminergiques/métabolisme , Agonistes histaminergiques/pharmacologie , Mâle , Souris , Facteurs de protection , ARN messager/métabolisme , Ranitidine/métabolisme , Ranitidine/pharmacologie , Récepteur histaminergique H2/génétique , Récepteur histaminergique H2/métabolisme , Thiazoles
9.
PLoS One ; 17(9): e0274297, 2022.
Article de Anglais | MEDLINE | ID: mdl-36099304

RÉSUMÉ

The liver is the main organ that regulates lipid and glucose metabolism. Ectopic lipid accumulation in the liver impairs insulin sensitivity and glucose metabolism. Lipoprotein lipase (LPL), mainly expressed in the adipose tissue and muscle, is a key enzyme that regulates lipid metabolism via the hydrolysis of triglyceride in chylomicrons and very-low-density lipoproteins. Here, we aimed to investigate whether the suppression level of hepatic lipid accumulation via overexpression of LPL in mouse liver leads to improved metabolism. To overexpress LPL in the liver, we generated an LPL-expressing adenovirus (Ad) vector using an improved Ad vector that exhibited considerably lower hepatotoxicity (Ad-LPL). C57BL/6 mice were treated with Ad vectors and simultaneously fed a high-fat diet (HFD). Lipid droplet formation in the liver decreased in Ad-LPL-treated mice relative to that in control Ad vector-treated mice. Glucose tolerance and insulin resistance were remarkably improved in Ad-LPL-treated mice compared to those in control Ad vector-treated mice. The expression levels of fatty acid oxidation-related genes, such as peroxisome proliferator-activated receptor α, carnitine palmitoyltransferase 1, and acyl-CoA oxidase 1, were 1.7-2.0-fold higher in Ad-LPL-treated mouse livers than that in control Ad-vector-treated mouse livers. Furthermore, hepatic LPL overexpression partly maintained mitochondrial content in HFD-fed mice. These results indicate that LPL overexpression in the livers of HFD-fed mice attenuates the accumulation of lipid droplets in the liver and improves glucose metabolism. These findings may enable the development of new drugs to treat metabolic syndromes such as type 2 diabetes mellitus and non-alcoholic fatty liver disease.


Sujet(s)
Diabète de type 2 , Insulinorésistance , Animaux , Diabète de type 2/métabolisme , Alimentation riche en graisse , Glucose/métabolisme , Insulinorésistance/physiologie , Lipoprotein lipase/génétique , Lipoprotein lipase/métabolisme , Foie/métabolisme , Souris , Souris de lignée C57BL , Triglycéride/métabolisme
10.
Exp Clin Endocrinol Diabetes ; 130(4): 254-261, 2022 Apr.
Article de Anglais | MEDLINE | ID: mdl-33782927

RÉSUMÉ

Genome-wide association studies have identified more than 300 loci associated with type 2 diabetes mellitus; however, the mechanisms underlying their role in type 2 diabetes mellitus susceptibility remain largely unknown. Zinc finger AN1-type domain 3 (ZFAND3), known as testis-expressed sequence 27, is a type 2 diabetes mellitus-susceptibility gene. Limited information is available regarding the physiological role of ZFAND3 in vivo. This study aimed to investigate the association between ZFAND3 and type 2 diabetes mellitus. ZFAND3 was significantly upregulated in the liver of diabetic mice compared to wild-type mice. To overexpress ZFAND3, we generated a ZFAND3-expressing adenovirus (Ad) vector using an improved Ad vector exhibiting significantly lower hepatotoxicity (Ad-ZFAND3). Glucose tolerance was significantly improved in Ad-ZFAND3-treated mice compared to the control Ad-treated mice. ZFAND3 overexpression in the mouse liver also improved insulin resistance. Furthermore, gluconeogenic gene expression was significantly lower in primary mouse hepatocytes transduced with Ad-ZFAND3 than those transduced with the control Ad vector. The present results suggest that ZFAND3 improves glucose tolerance by improving insulin resistance and suppressing gluconeogenesis, serving as a potential novel therapeutic target for type 2 diabetes mellitus.


Sujet(s)
Diabète expérimental , Diabète de type 2 , Insulinorésistance , Animaux , Diabète expérimental/métabolisme , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Diabète de type 2/thérapie , Étude d'association pangénomique , Glucose/métabolisme , Insulinorésistance/génétique , Foie/métabolisme , Mâle , Souris
11.
Biol Pharm Bull ; 44(11): 1759-1766, 2021.
Article de Anglais | MEDLINE | ID: mdl-34719652

RÉSUMÉ

Vasogenic edema results from blood-brain barrier (BBB) disruption after traumatic brain injury (TBI), and although it can be fatal, no promising therapeutic drugs have been developed as yet. Transient receptor potential vanilloid 4 (TRPV4) is a calcium-permeable channel that is sensitive to temperature and osmotic pressure. As TRPV4 is known to be responsible for various pathological conditions following brain injury, we investigated the effects of pharmacological TRPV4 antagonists on TBI-induced vasogenic edema in this study. A TBI model was established by inflicting fluid percussion injury (FPI) in the mouse cerebrum and cultured astrocytes. Vasogenic brain edema and BBB disruption were assessed based on brain water content and Evans blue (EB) extravasation into brain tissue, respectively. After FPI, brain water content and EB extravasation increased. Repeated intracerebroventricular administration of the specific TRPV4 antagonists HC-067047 and RN-1734 dose-dependently reduced brain water content and alleviated EB extravasation in FPI mice. Additionally, real-time PCR analysis indicated that administration of HC-067047 and RN-1734 reversed the FPI-induced increase in mRNA levels of endogenous causal factors for BBB disruption, including matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor-A (VEGF-A), and endothelin-1 (ET-1). In astrocytes, TRPV4 level was observed to be higher than that in brain microvascular endothelial cells. Treatment with HC-067047 and RN-1734 inhibited the increase in mRNA levels of MMP-9, VEGF-A, and ET-1 in cultured astrocytes subjected to in vitro FPI. These results suggest that pharmacological inhibition of TRPV4 is expected to be a promising therapeutic strategy for treating TBI-induced vasogenic edema.


Sujet(s)
Oedème cérébral/traitement médicamenteux , Lésions traumatiques de l'encéphale/traitement médicamenteux , Canaux cationiques TRPV/antagonistes et inhibiteurs , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Oedème cérébral/étiologie , Lésions traumatiques de l'encéphale/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Matrix metalloproteinase 9/métabolisme , Souris , Morpholines/pharmacologie , Pyrroles/pharmacologie , Réaction de polymérisation en chaine en temps réel , Sulfonamides/pharmacologie , Canaux cationiques TRPV/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme
12.
Int J Mol Sci ; 22(12)2021 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-34203960

RÉSUMÉ

Traumatic brain injury (TBI) is immediate damage caused by a blow to the head resulting from traffic accidents, falls, and sporting activity, which causes death or serious disabilities in survivors. TBI induces multiple secondary injuries, including neuroinflammation, disruption of the blood-brain barrier (BBB), and brain edema. Despite these emergent conditions, current therapies for TBI are limited or insufficient in some cases. Although several candidate drugs exerted beneficial effects in TBI animal models, most of them failed to show significant effects in clinical trials. Multiple studies have suggested that astrocytes play a key role in the pathogenesis of TBI. Increased reactive astrocytes and astrocyte-derived factors are commonly observed in both TBI patients and experimental animal models. Astrocytes have beneficial and detrimental effects on TBI, including promotion and restriction of neurogenesis and synaptogenesis, acceleration and suppression of neuroinflammation, and disruption and repair of the BBB via multiple bioactive factors. Additionally, astrocytic aquaporin-4 is involved in the formation of cytotoxic edema. Thus, astrocytes are attractive targets for novel therapeutic drugs for TBI, although astrocyte-targeting drugs have not yet been developed. This article reviews recent observations of the roles of astrocytes and expected astrocyte-targeting drugs in TBI.


Sujet(s)
Astrocytes/anatomopathologie , Lésions traumatiques de l'encéphale/anatomopathologie , Lésions traumatiques de l'encéphale/physiopathologie , Animaux , Barrière hémato-encéphalique/anatomopathologie , Humains , Néovascularisation physiologique , Neurogenèse , Transduction du signal
13.
Neurochem Int ; 146: 105042, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33838160

RÉSUMÉ

In the adult brain, sonic hedgehog acts on cerebral microvascular endothelial cells to stabilize the blood-brain barrier. The expression of sonic hedgehog by astrocytes is altered during brain injury, and this change has been shown to affect permeability of blood-brain barrier. However, much remains unknown about the regulation of astrocytic sonic hedgehog production. Our results showed that endothelin-1 reduced sonic hedgehog mRNA expression and extracellular protein release in mouse cerebral cultured astrocytes, but had no effect in bEnd.3, a mouse brain microvascular endothelial-derived cell line. The effect of endothelin-1 on astrocyte sonic hedgehog expression was suppressed by an ETB antagonist BQ788, but was unchanged by the ETA antagonist FR139317. In cultured astrocytes and bEnd.3, endothelin-1 did not affect the expression of the sonic hedgehog receptor-related molecules, patched-1 and smoothened. In an animal model of traumatic brain injury, fluid percussion injury on the mouse cerebrum increased the expression of sonic hedgehog, patched-1, and smoothened. Repeated administration of BQ788 enhanced sonic hedgehog expression at 5 days after fluid percussion injury. Histochemical examination revealed sonic hedgehog expression in glial fibrillary acidic protein-positive astrocytes in the cerebrum after fluid percussion injury. Administration of exogenous sonic hedgehog and BQ788 suppressed Evans blue extravasation, an indicator of blood vessel permeability, induced by fluid percussion injury. The effects of BQ788 on fluid percussion injury-induced Evans blue extravasation were reduced by the administration of jervine, a sonic hedgehog inhibitor. Altogether, these results suggest that endothelin-1 down-regulates astrocytic sonic hedgehog to promote disruption of the blood-brain barrier during traumatic brain injury.


Sujet(s)
Astrocytes/métabolisme , Barrière hémato-encéphalique/métabolisme , Lésions traumatiques de l'encéphale/métabolisme , Endothéline-1/pharmacologie , Protéines Hedgehog/métabolisme , Récepteur de l'endothéline de type B/métabolisme , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Lésions traumatiques de l'encéphale/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/physiologie , Protéines Hedgehog/antagonistes et inhibiteurs , Mâle , Souris , Oligopeptides/pharmacologie , Pipéridines/pharmacologie , Alcaloïdes de Veratrum/pharmacologie
14.
Neuropharmacology ; 175: 108182, 2020 09 15.
Article de Anglais | MEDLINE | ID: mdl-32561219

RÉSUMÉ

Traumatic brain injury (TBI) is induced by the immediate physical disruption of brain tissue. TBI causes disruption of the blood-brain barrier (BBB) and brain edema. In the cerebrospinal fluid (CSF) of TBI patients, endothelin-1 (ET-1) is increased, suggesting that ET-1 aggravates TBI-induced brain damage. In this study, the effect of bosentan (ETA/ETB antagonist) and ambrisentan (ETA antagonist) on BBB dysfunction and brain edema were examined in a mouse model of TBI using lateral fluid percussion injury (FPI). FPI to the mouse cerebrum increased the expression levels of ET-1 and ETB receptors. Administration of bosentan (3 or 15 mg/kg/day) and ambrisentan (0.1 or 0.5 mg/kg/day) at 6 and 24 h after FPI ameliorated BBB disruption and cerebral brain edema. Delayed administration of bosentan from 2 days after FPI also reduced BBB disruption and brain edema, while ambrisentan had no significant effects. FPI-induced expression levels of ET-1 and ETB receptors were reduced by bosentan, but not by ambrisentan. In cultured mouse astrocytes and brain microvessel endothelial cells, ET-1 (100 nM) increased prepro--ET-1 mRNA, which was inhibited by bosentan, but not by ambrisentan. FPI-induced alterations of the expression levels of matrix metalloproteinase-9, vascular endothelial growth factor-A, and angiopoietin-1 in the mouse cerebrum were reduced by delayed administration of bosentan, while ambrisentan had no significant effects. These results suggest that ET antagonists are effective in improving BBB disruption and cerebral edema in TBI patients and that an ETA/ETB non-selective type of antagonists is more effective.


Sujet(s)
Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Bosentan/administration et posologie , Oedème cérébral/métabolisme , Lésions traumatiques de l'encéphale/métabolisme , Antagonistes des récepteurs de l'endothéline/administration et posologie , Phénylpropionates/administration et posologie , Pyridazines/administration et posologie , Animaux , Oedème cérébral/complications , Lésions traumatiques de l'encéphale/complications , Antagonistes du récepteur de type B de l'endothéline , Endothéline-1/antagonistes et inhibiteurs , Endothéline-1/métabolisme , Mâle , Souris , Récepteur de l'endothéline de type B/administration et posologie , Récepteur de l'endothéline de type B/métabolisme
15.
J Neurochem ; 154(3): 330-348, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-31957020

RÉSUMÉ

Angiopoietin-1, an angiogenic factor, stabilizes brain microvessels through Tie-2 receptor tyrosine kinase. In traumatic brain injury, blood-brain barrier (BBB) disruption is an aggravating factor that induces brain edema and neuroinflammation. We previously showed that BQ788, an endothelin ETB receptor antagonist, promoted recovery of BBB function after lateral fluid percussion injury (FPI) in mice. To clarify the mechanisms underlying BBB recovery mediated by BQ788, we examined the involvements of the angiopoietin-1/Tie-2 signal. When angiopoietin-1 production and Tie-2 phosphorylation were assayed by quantitative reverse transcription polymerase chain reaction and western blotting, increased angiopoietin-1 production and Tie-2 phosphorylation were observed in 7-10 days after FPI in the mouse cerebrum, whereas no significant effects were obtained at 5 days. When BQ788 (15 nmol/day, i.c.v.) were administered in 2-5 days after FPI, increased angiopoietin-1 production and Tie-2 phosphorylation were observed. Immunohistochemical observations showed that brain microvessels and astrocytes contained angiopoietin-1 after FPI, and brain microvessels also contained phosphorylated Tie-2. Treatment with endothelin-1 (100 nM) decreased angiopoietin-1 production in cultured astrocytes and the effect was inhibited by BQ788 (1 µM). Five days after FPI, increased extravasation of Evans blue dye accompanied by reduction in claudin-5, occludin, and zonula occludens-1 proteins were observed in mouse cerebrum while these effects of FPI were reduced by BQ788 and exogenous angiopoietin-1 (1 µg/day, i.c.v.). The effects of BQ788 were inhibited by co-administration of a Tie-2 kinase inhibitor (40 nmol/day, i.c.v.). These results suggest that BQ788 administration after traumatic brain injury promotes recovery of BBB function through activation of the angiopoietin-1/Tie-2 signal.


Sujet(s)
Angiopoïétine-1/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Lésions traumatiques de l'encéphale/métabolisme , Antagonistes du récepteur de type B de l'endothéline/pharmacologie , Oligopeptides/pharmacologie , Pipéridines/pharmacologie , Récepteur TIE-2/métabolisme , Animaux , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Cerveau/effets des médicaments et des substances chimiques , Cerveau/traumatismes , Cerveau/métabolisme , Mâle , Souris
16.
Int J Mol Sci ; 20(3)2019 Jan 29.
Article de Anglais | MEDLINE | ID: mdl-30699952

RÉSUMÉ

The blood-brain barrier (BBB) is a major functional barrier in the central nervous system (CNS), and inhibits the extravasation of intravascular contents and transports various essential nutrients between the blood and the brain. After brain damage by traumatic brain injury, cerebral ischemia and several other CNS disorders, the functions of the BBB are disrupted, resulting in severe secondary damage including brain edema and inflammatory injury. Therefore, BBB protection and recovery are considered novel therapeutic strategies for reducing brain damage. Emerging evidence suggests key roles of astrocyte-derived factors in BBB disruption and recovery after brain damage. The astrocyte-derived vascular permeability factors include vascular endothelial growth factors, matrix metalloproteinases, nitric oxide, glutamate and endothelin-1, which enhance BBB permeability leading to BBB disruption. By contrast, the astrocyte-derived protective factors include angiopoietin-1, sonic hedgehog, glial-derived neurotrophic factor, retinoic acid and insulin-like growth factor-1 and apolipoprotein E which attenuate BBB permeability resulting in recovery of BBB function. In this review, the roles of these astrocyte-derived factors in BBB function are summarized, and their significance as therapeutic targets for BBB protection and recovery after brain damage are discussed.


Sujet(s)
Astrocytes/métabolisme , Barrière hémato-encéphalique/métabolisme , Lésions encéphaliques/métabolisme , Animaux , Humains , Jonctions serrées/métabolisme
17.
J Biol Chem ; 294(11): 3920-3933, 2019 03 15.
Article de Anglais | MEDLINE | ID: mdl-30670587

RÉSUMÉ

Brain injury-mediated induction of reactive astrocytes often leads to glial scar formation in damaged brain regions. Activation of signal transducer and activator of transcription 3 (STAT3), a member of the STAT family of transcription factors, plays a pivotal role in inducing reactive astrocytes and glial scar formation. Endothelin-1 (ET-1) is a vasoconstrictor peptide, and its levels increase in brain disorders and promote astrocytic proliferation through ETB receptors. To clarify the mechanisms underlying ET-1-mediated astrocytic proliferation, here we examined its effects on STAT3 in cultured rat astrocytes. ET-1 treatment stimulated Ser-727 phosphorylation of STAT3 in the astrocytes, but Tyr-705 phosphorylation was unaffected, and ET-induced STAT3 Ser-727 phosphorylation was reduced by the ETB antagonist BQ788. ET-1 stimulated STAT3 binding to its consensus DNA-binding motifs. Monitoring G1/S phase cell cycle transition through bromodeoxyuridine (BrdU) incorporation, we found that ET-1 increases BrdU incorporation into the astrocytic nucleus, indicating cell cycle progression. Of note, STAT3 chemical inhibition (with stattic or 5,15-diphenyl-porphine (5,15-DPP)) or siRNA-mediated STAT3 silencing reduced ET-induced BrdU incorporation. Moreover, ET-1 increased astrocytic expression levels of cyclin D1 and S-phase kinase-associated protein 2 (SKP2), which were reduced by stattic, 5,15-DPP, and STAT3 siRNA. ChIP-based PCR analysis revealed that ET-1 promotes the binding of SAT3 to the 5'-flanking regions of rat cyclin D1 and SKP2 genes. Our results suggest that STAT3-mediated regulation of cyclin D1 and SKP2 expression underlies ET-induced astrocytic proliferation.


Sujet(s)
Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Cycline D1/métabolisme , Endothéline-1/pharmacologie , Protéines associées aux kinases de la phase S/métabolisme , Facteur de transcription STAT-3/métabolisme , Animaux , Astrocytes/cytologie , Astrocytes/enzymologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cycline D1/génétique , Relation dose-effet des médicaments , Phosphorylation/effets des médicaments et des substances chimiques , Petit ARN interférent/pharmacologie , Rats , Rat Wistar , Protéines associées aux kinases de la phase S/génétique , Facteur de transcription STAT-3/antagonistes et inhibiteurs , Relation structure-activité
18.
J Pharmacol Sci ; 138(1): 54-62, 2018 Sep.
Article de Anglais | MEDLINE | ID: mdl-30301597

RÉSUMÉ

Narrowband-ultraviolet B (NB-UVB) phototherapy is used for the treatment of atopic dermatitis. Previously, we reported that irradiation with 200 mJ/cm2 of 310 nm NB-UVB suppressed phorbol-12-myristate-13-acetate (PMA)-induced up-regulation of histamine H1 receptor (H1R) gene expression without induction of apoptosis in HeLa cells. However, the effect of NB-UVB irradiation on nasal symptoms is still unclear. Here, we show that low dose irradiation with 310 nm NB-UVB alleviates nasal symptoms in toluene 2,4-diisocyanate (TDI)-sensitized allergy model rats. Irradiation with 310 nm NB-UVB suppressed PMA-induced H1R mRNA up-regulation in HeLa cells dose-dependently at doses of 75-200 mJ/cm2 and reversibly at a dose of 150 mJ/cm2 without induction of apoptosis. While, at doses of more than 200 mJ/cm2, irradiation with 310 nm NB-UVB induced apoptosis. Western blot analysis showed that the suppressive effect of NB-UVB irradiation on H1R gene expression was through the inhibition of ERK phosphorylation. In TDI-sensitized rat, intranasal irradiation with 310 nm NB-UVB at an estimated dose of 100 mJ/cm2 once a day for three days suppressed TDI-induced sneezes and up-regulation of H1R mRNA in nasal mucosa without induction of apoptosis. These findings suggest that repeated intranasal irradiation with low dose of NB-UVB could be clinically used as phototherapy of AR.


Sujet(s)
Apoptose/effets des radiations , Expression des gènes/effets des radiations , Muqueuse nasale/anatomopathologie , Muqueuse nasale/effets des radiations , ARN messager/métabolisme , Récepteur histaminergique H1/génétique , Récepteur histaminergique H1/métabolisme , Rayons ultraviolets , Régulation positive/effets des radiations , Animaux , Relation dose-effet des rayonnements , Cellules HeLa , Humains , Mâle , Photothérapie , Rats , Rhinite allergique/thérapie
19.
J Neurotrauma ; 35(13): 1481-1494, 2018 07 01.
Article de Anglais | MEDLINE | ID: mdl-29316834

RÉSUMÉ

Traumatic brain injury (TBI) is induced by immediate physical disruption of brain tissue, and causes death and disability. Studies on experimental TBI animal models show that disruption of the blood-brain barrier (BBB) underlies brain edema and neuroinflammation during the delayed phase of TBI. In neurological disorders, endothelin-1 (ET-1) is involved in BBB dysfunction and brain edema. In this study, the effect of ET antagonists on BBB dysfunction and brain edema were examined in a mouse focal TBI model using lateral fluid percussion injury (FPI). ET-1 and ETB receptors were increased at 2-7 days after FPI, which was accompanied by extravasation of Evans blue (EB) and brain edema. Repeated intracerebroventricular administration of BQ788 (15 nmol/day), an ETB antagonist, from 2 days after FPI promoted recovery of EB extravasation and brain edema, while FR 139317, an ETA antagonist, had no effect. Delayed intravenous administration of BQ788 also promoted recovery from FPI-induced EB extravasation and brain edema. While FPI caused decreases in claudin-5, occludin, and zonula occludens-1 proteins, BQ788 reversed FPI-induced reductions of them. Immunohistochemical observation of the cerebrum after FPI showed that ETB receptors are predominantly expressed in glial fibrillary acidic protein (GFAP)-positive astrocytes. BQ788 reduced FPI-induced increases in GFAP-positive astrocytes. GFAP-positive astrocytes produced vascular endothelial growth factor-A (VEGF-A) and matrix metalloproteinase-9 (MMP9). FPI-induced increases in VEGF-A and MMP-9 production were reversed by BQ788. These results suggest that ETB receptor antagonism during the delayed phase of focal TBI promotes recovery of BBB function and reduction of brain edema.


Sujet(s)
Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Oedème cérébral/étiologie , Lésions traumatiques de l'encéphale/complications , Antagonistes du récepteur de type B de l'endothéline/pharmacologie , Oligopeptides/pharmacologie , Pipéridines/pharmacologie , Animaux , Oedème cérébral/anatomopathologie , Lésions traumatiques de l'encéphale/anatomopathologie , Modèles animaux de maladie humaine , Injections ventriculaires , Mâle , Souris
20.
Yakugaku Zasshi ; 137(10): 1241-1246, 2017.
Article de Japonais | MEDLINE | ID: mdl-28966265

RÉSUMÉ

Brain edema is a severe morbid complication of brain injury, characterized by excessive fluid accumulation and an elevation of intracranial pressure. However, effective anti-brain edema drugs are lacking. One of the causes of brain edema is disruption of blood-brain barrier (BBB) function, which results in extravasation of intravascular fluid. After brain damage, astrocytes are activated, and astrocyte-derived vascular endothelial growth factor-A (VEGF-A) is known to induce BBB dysfunction. Therefore maintaining BBB integrity by regulating astrocyte function is a potentially effective strategy for treating brain edema. In this review, we focus on the endothelin ETB receptor and its role in regulation of astrocyte functions. In mice, brain damage was induced by fluid percussion injury (FPI), and the resulting BBB disruption and brain edema were observed in the mouse cerebrum. BQ788, a selective ETB receptor antagonist, attenuated the FPI-induced BBB disruption and brain edema. Levels of brain VEGF-A increased after FPI, mainly in reactive astrocytes. BQ788 suppressed the FPI-induced increase in VEGF-A expression in reactive astrocytes. Moreover, intraventricular administration of VEGF neutralizing antibody also attenuated FPI-induced BBB disruption and brain edema. Claudin-5 is an endothelial tight junction protein essential for normal BBB structure and function. Levels of claudin-5 protein were reduced by FPI. Furthermore, VEGF neutralizing antibody blocked FPI-induced decrease in claudin-5. These results suggest that the ETB receptor antagonist BQ788 protects against brain edema by inhibiting VEGF-A-mediated decrease in claudin-5.


Sujet(s)
Astrocytes/physiologie , Oedème cérébral/étiologie , Oedème cérébral/prévention et contrôle , Antagonistes du récepteur de type B de l'endothéline , Oligopeptides/pharmacologie , Oligopeptides/usage thérapeutique , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Récepteur de l'endothéline de type B/physiologie , Facteur de croissance endothéliale vasculaire de type A/physiologie , Animaux , Barrière hémato-encéphalique/physiologie , Barrière hémato-encéphalique/physiopathologie , Encéphale/métabolisme , Oedème cérébral/métabolisme , Claudine-5/métabolisme , Claudine-5/physiologie , Modèles animaux de maladie humaine , Expression des gènes/effets des médicaments et des substances chimiques , Souris , Protéines de la jonction serrée/métabolisme , Protéines de la jonction serrée/physiologie , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE