Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
3.
Neuroscience ; 289: 214-23, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25592431

ABSTRACT

Diabetes mellitus (DM) is a major risk factor for stroke and it exacerbates tissue damage after ischemic insult. Diabetes is one of the important causes of the progression of white matter lesion, however, the pathological mechanisms remain unclear. The present study evaluated the influences of type 2 DM on ischemic subcortical white matter injury and the recruitment of oligodendrocyte progenitor cells (OPCs) under chronic cerebral hypoperfusion using type 2 diabetic (db/db) mice. After bilateral common carotid artery stenosis (BCAS), the rarefaction in the white matter was more severe in db/db mice than in db/+ mice, and the number of glutathione S-transferase-pi (GST-pi)-positive mature oligodendrocytes (OLG) was lower in db/db mice than in db/+ mice at 4 and 8 weeks after ischemia. There were no significant differences in the number of single-stranded DNA (ssDNA)-positive apoptotic cells in the deep white matter between the db/db and db/+ mice. We found a transient increase in the platelet-derived growth factor receptor-α (PDGFRα)-positive OPCs in white matter lesions after ischemia. However, significantly fewer PDGFRα-positive OPCs were detected in db/db than db/+ mice from 4weeks after BCAS. The number of Ki67-positive proliferating cells in the deep white matter was significantly lower in db/db mice than in db/+ mice from 4 to 8weeks after BCAS. Most of the Ki67-positive cells were PDGFRα-positive OPCs. Finally, we assessed the survival of 5-bromo-2'-deoxyuridine (BrdU)-positive proliferating cells in ischemic white matter, and found significantly poorer survival of BrdU/PDGFRα-positive OPCs or BrdU/GST-pi-positive OLGs in the db/db mice compared to the db/+ mice in the white matter after BCAS. Our findings suggest that the type 2 DM mice exhibited more severe white matter injury 8 weeks after chronic ischemia. Decreased proliferation and survival of OPCs may play an important role in the progression of white matter lesions after ischemia in diabetics.


Subject(s)
Brain Ischemia/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Neural Stem Cells/physiology , Oligodendroglia/physiology , White Matter/physiopathology , Animals , Apoptosis/physiology , Brain/pathology , Brain/physiopathology , Brain Ischemia/pathology , Carotid Stenosis , Cell Proliferation/physiology , Cell Survival/physiology , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Glutathione S-Transferase pi/metabolism , Ki-67 Antigen/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Neural Stem Cells/pathology , Oligodendroglia/pathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , White Matter/pathology
4.
Neuroscience ; 278: 113-21, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25135354

ABSTRACT

Heat shock protein 27 (HSP27) exerts cytoprotection against many cellular insults including cerebral ischemia. We previously indicated that intravenous injection of HSP27 purified from human lymphocytes (hHSP27) significantly reduced infarct volume following cerebral ischemia-reperfusion injury, while recombinant HSP27 (rHSP27) was less effective. Phosphorylation is important for HSP27 function, and hHSP27 was more highly phosphorylated than rHSP27. We hypothesized that MAPKAP kinase 2 in vitro-phosphorylated rHSP27 (prHSP27) might increase its brain protection. Mice underwent transient 1-h middle cerebral artery occlusion (MCAO), and then received tail-vein injections of one of the following 1h after reperfusion: hHSP27 as positive control, rHSP27, prHSP27, or bovine serum albumin (BSA) as control. We measured infarct volume, neurological deficits, neurological severity, physiological parameters, cell-death, oxidative stress, and inflammatory response. Compared with BSA controls (30.7±3.1mm(3), n=5), infarct volume was reduced by 67% in the hHSP27 positive-control group (10.1±4.6mm(3), P<0.001, n=5), 17% following rHSP27 (25.4±3.6mm(3), P<0.05, n=5), and 46% following prHSP27 (16.5±4.0mm(3), P<0.001, n=9). Compared to the rHSP27 and BSA-treated groups, prHSP27 also reduced functional deficits, and significantly suppressed apoptosis, oxidative stress, and inflammatory responses. Here, we showed the superior neuroprotective effects of phosphorylated HSP27 by administering prHSP27. prHSP27 may be a useful therapeutic agent to protect against acute cerebral ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , HSP27 Heat-Shock Proteins/administration & dosage , HSP27 Heat-Shock Proteins/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/pathology , Cell Death/drug effects , Encephalitis/metabolism , Humans , Infarction, Middle Cerebral Artery , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism
5.
Eur J Neurol ; 21(4): 667-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24495037

ABSTRACT

BACKGROUND AND PURPOSE: Obesity is associated with the risk of coronary artery disease and stroke. Visceral fat plays a significant role in the atherogenic effects of obesity. Whether visceral fat accumulation, as measured by computed tomography (CT), is an independent risk factor for the presence of cerebral small vessel disease (SVD) was investigated. METHODS: This study comprised 506 Japanese subjects 35-74 years of age (mean 55.3 years) without a history of symptomatic cerebrovascular disease who underwent health screening tests, including brain magnetic resonance imaging, carotid echography and measurements of the visceral fat area (VFA) and subcutaneous fat area (SFA) on abdominal CT. Visceral fat accumulation was defined as VFA ≥ 100 cm(2) . Logistic regression analysis was performed to examine the associations between visceral fat accumulation and cerebral SVD such as white matter lesions (WMLs) and silent lacunar infarction (SLI). RESULTS: The prevalence of WMLs and SLI but not carotid plaque were significantly higher in subjects with VFA ≥ 100 cm(2) than those with VFA < 100 cm(2) . A VFA ≥ 100 cm(2) was associated with WMLs and SLI independent of age, cardiovascular risk factors and other measurements of obesity, such as waist circumference and body mass index. A large waist circumference was independently associated with SLI. SFA, the combination of VFA and SFA, and body mass index were not associated with WMLs or SLI. CONCLUSIONS: Visceral fat accumulation was independently associated with the presence of cerebral SVD in subjects without a history of symptomatic cerebrovascular disease.


Subject(s)
Cerebral Small Vessel Diseases/etiology , Cerebral Small Vessel Diseases/pathology , Intra-Abdominal Fat/pathology , Adult , Aged , Brain/pathology , Female , Humans , Intra-Abdominal Fat/metabolism , Japan , Logistic Models , Magnetic Resonance Imaging , Male , Middle Aged , Retrospective Studies , Risk Management , Tomography, X-Ray Computed , Ultrasonography
6.
Neuroscience ; 244: 113-21, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23602887

ABSTRACT

Glutamate plays a central role in brain physiology and pathology. The involvement of excitatory amino acid transporters (EAATs) in neurodegenerative disorders including acute stroke has been widely studied, but little is known about the role of glial glutamate transporters in white matter injury after chronic cerebral hypoperfusion. The present study evaluated the expression of glial (EAAT1 and EAAT2) and neuronal (EAAT3) glutamate transporters in subcortical white matter and cortex, before and 3-28 days after the ligation of bilateral common carotid arteries (LBCCA) in rat brain. K-B staining showed a gradual increase of demyelination in white matter after ischemia, while there was no cortical involvement. Between 3 and 7 days after LBCCA, a significant increase in EAAT2 protein levels was observed in the ischemic brain and the number of EAAT2-positive cells also significantly increased both in the cortical and white matter lesions. EAAT2 was detected in glial-fibrillary acidic protein (GFAP)-positive astrocytes in both the cortex and white matter, but not in neuronal and oligodendroglial cells. EAAT1 was slightly elevated after ischemia only in the white matter, but EAAT3 was at almost similar levels both in the cortex and white matter after ischemia. A significant increase in EAAT2 expression level was also noted in the deep white matter of chronic human ischemic brain tissue compared to the control group. Our findings suggest important roles for up-regulated EAAT2 in chronic brain ischemia especially in the regulation of high-affinity of extracellular glutamate and minimization of white matter damage.


Subject(s)
Brain Ischemia/metabolism , Corpus Callosum/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Glutamate Plasma Membrane Transport Proteins/biosynthesis , Nerve Fibers, Myelinated/metabolism , Adult , Aged , Aged, 80 and over , Animals , Astrocytes/metabolism , Brain Ischemia/complications , Brain Ischemia/pathology , Case-Control Studies , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chronic Disease , Corpus Callosum/pathology , Demyelinating Diseases/complications , Demyelinating Diseases/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 3/biosynthesis , Female , Humans , Male , Nerve Fibers, Myelinated/pathology , Neurons/metabolism , Oligodendroglia/metabolism , Rats , Up-Regulation
7.
J Comp Pathol ; 148(4): 410-3, 2013 May.
Article in English | MEDLINE | ID: mdl-22980777

ABSTRACT

This report describes an outbreak of yersiniosis in Egyptian rousette bats (Rousettus aegyptiacus) caused by Yersinia pseudotuberculosis serotype 4b. Twelve of 61 bats died between November and December 2008 or in May 2009. The bats often displayed multiple yellow-white nodules in the spleen and liver. Microscopically, these consisted of focal necrosis accompanied by inflammatory cell infiltration and colonies of gram-negative bacilli. The bacterial colonies were identified immunohistochemically as Y. pseudotuberculosis O4 and Y. pseudotuberculosis serotype 4b was identified by bacteriological examination. Polymerase chain reaction demonstrated that the isolate harboured the virulence genes virF, inv and ypmA. YPMa is as a superantigenic toxin that is associated with acute systemic infection in man and may contribute to the virulence of Y. pseudotuberculosis in bats.


Subject(s)
Chiroptera/microbiology , Liver/pathology , Spleen/pathology , Yersinia pseudotuberculosis Infections/veterinary , Yersinia pseudotuberculosis/isolation & purification , Animals , Disease Outbreaks , Liver/microbiology , Male , Spleen/microbiology , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis Infections/pathology
8.
Neuroscience ; 171(4): 1367-76, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-20933581

ABSTRACT

Evidence suggests that neurogenesis occurs in the adult mammalian brain, and that various stimuli, for example, ischemia/hypoxia, enhance the generation of neural progenitor cells in the subventricular zone (SVZ) and their migration into the olfactory bulb. In a mouse stroke model, focal ischemia results in activation of neural progenitor cells followed by their migration into the ischemic lesion. The present study assessed the in vivo effects of cilostazol, a type 3 phosphodiesterase inhibitor known to activate the cAMP-responsive element binding protein (CREB) signaling, on neurogenesis in the ipsilateral SVZ and peri-infarct area in a mouse model of transient middle cerebral artery occlusion. Mice were divided into sham operated (n=12), vehicle- (n=18) and cilostazol-treated (n=18) groups. Sections stained for 5-bromodeoxyuridine (BrdU) and several neuronal and a glial markers were analyzed at post-ischemia days 1, 3 and 7. Cilostazol reduced brain ischemic volume (P<0.05) and induced earlier recovery of neurologic deficit (P<0.05). Cilostazol significantly increased the density of BrdU-positive newly-formed cells in the SVZ compared with the vehicle group without ischemia. Increased density of doublecortin (DCX)-positive and BrdU/DCX-double positive neural progenitor cells was noted in the ipsilateral SVZ and peri-infarct area at 3 and 7 days after focal ischemia compared with the vehicle group (P<0.05). Cilostazol increased DCX-positive phosphorylated CREB (pCREB)-expressing neural progenitor cells, and increased brain derived neurotrophic factor (BDNF)-expressing astrocytes in the ipsilateral SVZ and peri-infarct area. The results indicated that cilostazol enhanced neural progenitor cell generation in both ipsilateral SVZ and peri-infarct area through CREB-mediated signaling pathway after focal ischemia.


Subject(s)
Cerebral Ventricles/physiopathology , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Neurogenesis/drug effects , Neuroprotective Agents/therapeutic use , Tetrazoles/therapeutic use , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Bromodeoxyuridine/metabolism , CREB-Binding Protein/metabolism , Cell Count/methods , Cerebral Ventricles/drug effects , Cilostazol , Disease Models, Animal , Doublecortin Protein , In Situ Nick-End Labeling/methods , Ischemic Attack, Transient/complications , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurologic Examination/methods , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Regional Blood Flow/drug effects
9.
Neuroscience ; 163(1): 296-307, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19531375

ABSTRACT

Cerebral ischemia/reperfusion injury is characterized by the development of inflammatory response, in which vascular macrophages and endogenous microglia are involved. Recent studies showed marked induction of hematopoietic prostaglandin D synthase (HPGDS) after ischemic/reperfusion injury and its localization in microglia, but the molecular mechanism(s) of HPGDS actions in cerebral ischemia is not clear. To clarify the role of HPGDS in cerebral ischemia, C57BL/6 mice and bone marrow chimera mice with cerebral ischemia/reperfusion injury were treated with (4-benzhydryloxy-(1) {3-(1H-tetrazol-5-yl)-propyl}piperidine (HQL-79), a specific inhibitor of HPGDS. The bone marrow chimera mice exhibit expression of enhanced green fluorescent protein (EGFP) in bone marrow/blood-derived monocytes/macrophages. Mice were subjected to ischemia/reperfusion and either treated with HQL-79 (n=44) or vehicle (n=44). Brain sections prepared at 72 h and 7 days after reperfusion were analyzed for neuronal nuclei (NeuN), HPGDS, ionized calcium-binding adapter molecule 1 (Iba1), inducible NO synthase (iNOS), nitrotyrosine, nuclear factor kappa B (NF-kB) and cyclooxygenase-2 (COX-2). The mortality rate (80%) and infarct size were larger in HQL-79- than vehicle-treated mice (58.7+/-8.5 versus 45.2+/-4.9 mm(3); mean+/-SEM, P<0.0001) at 7 days after reperfusion. HQL-79 reduced NeuN expression in the transition area and Iba1 expression (P<0.0001) in the ischemic peri- and penumbra area, but increased COX-2 (P<0.05) and NF-kB expression (P<0.05) in ischemic penumbra and increased formation of nitrotyrosine (P<0.0001) and iNOS (P<0.0001) in the ischemic core area at 72 h and 7 days after reperfusion. In EGFP chimera mice, HQL-79 increased the migration of Iba1/EGFP-positive bone marrow-derived monocytes/macrophages, and simultaneously upregulated iNOS expression in the ischemic core area (P<0.0001), but increased intrinsic microglia/macrophages in ischemic peri-area and penumbra (P<0.0001) at 72 h and 7 days after reperfusion, suggesting involvement of monocytes/macrophages in HQL-79-induced expansion of ischemic injury. Our results demonstrated that the neuroprotective effects of HPGDS in our model are mediated by suppression of activation and infiltration of inflammatory cells.


Subject(s)
Brain/drug effects , Encephalitis/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Intramolecular Oxidoreductases/metabolism , Ischemic Attack, Transient/drug therapy , Isomerases/metabolism , Lipocalins/metabolism , Reperfusion Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Biomarkers/metabolism , Bone Marrow Transplantation/methods , Brain/enzymology , Brain/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Disease Models, Animal , Encephalitis/physiopathology , Encephalitis/prevention & control , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Green Fluorescent Proteins/genetics , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/prevention & control , Intramolecular Oxidoreductases/antagonists & inhibitors , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/prevention & control , Isomerases/antagonists & inhibitors , Lipocalins/antagonists & inhibitors , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/physiology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type II/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Reperfusion Injury/physiopathology , Reperfusion Injury/prevention & control , Transplantation Chimera
10.
Neuroscience ; 162(2): 525-36, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19426786

ABSTRACT

Various stimuli, such as ischemia/hypoxia enhance newborn cell survival in the subventricular zone and their migration tangentially in chains toward the olfactory bulb. The present study assessed the fate of newborn neurons from subventricular zone to olfactory bulb under conditions of chronic cerebral hypoperfusion, and examined the role of cAMP-responsive element binding protein signaling on the survival of these neurons by using cilostazol, a potent inhibitor of type III phosphodiesterase. Rats underwent bilateral common carotid artery ligation. They were divided into sham-operated (n=70), vehicle- (n=70), and type III phosphodiesterase inhibitor-treated (n=70) groups. Immunohistochemically-stained section for 5-bromodeoxyuridine and a series of neuronal and glial markers were analyzed at days 7, 14, 21 and 28 after hypoperfusion. The reduction of olfactory bulb size gradually progressed in the vehicle group (P<0.05), but not in the sham-operated and type III phosphodiesterase inhibitor-treated group. The subventricular zone of the vehicle-treated rats contained significantly larger numbers of newborn neuroblasts after hypoperfusion, compared with sham-operated rats (P<0.05), but significantly lower numbers in the rostral migratory stream and olfactory bulb (P<0.05). Treatment of rats with type III phosphodiesterase inhibitor increased the number of neuroblasts and enhanced the survival and differentiation of cells (P<0.05). Phosphorylated cAMP-responsive element binding protein within neuroblasts was markedly decreased in the subventricular zone, rostral migratory stream, and olfactory bulb of vehicle-treated rats (P<0.05), but treatment with type III phosphodiesterase inhibitor resulted in recovery of this expression throughout hypoperfusion, leading to enhanced neurogenesis (P<0.05). These effects were abrogated by protein kinase A and C inhibitor. Our results indicated that cAMP-responsive element binding protein signaling is a key mediator of neurogenesis after prolonged hypoperfusion and provide the basis for new regenerative therapies for ischemic brain injury.


Subject(s)
Brain Ischemia/pathology , Cyclic AMP Response Element-Binding Protein/physiology , Neurons/pathology , Serine/metabolism , Stem Cells/pathology , Animals , Apoptosis , Brain Ischemia/etiology , Brain Ischemia/physiopathology , Carotid Stenosis/complications , Cerebral Ventricles/pathology , Cerebrovascular Circulation , Chronic Disease , Cilostazol , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Male , Olfactory Bulb/pathology , Phosphodiesterase 3 Inhibitors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Wistar , Signal Transduction , Tetrazoles/pharmacology
11.
Neuroscience ; 162(2): 317-27, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19409967

ABSTRACT

A multicenter randomized clinical trial demonstrated that acute ischemic stroke patients treated with edaravone, a scavenger of hydroxyl radicals, had significant functional improvement. We tested the hypothesis that edaravone has protective effects against white matter lesions (WML) and endothelial injury, using a rat chronic hypoperfusion model. Adult Wistar rats underwent ligation of bilateral common carotid artery (LBCCA) and were divided into the edaravone group (injected once only immediately after LBCCA [n=39, ED(1)]; and injected on three consecutive days [n=39, ED(3)]), the vehicle group (n=39), and the sham group (n=15). Cerebral blood flow, Morris water maze performance, footprint test for locomotor function, immunohistochemical analyses and Western blot analysis were performed before and after LBCCA. The ED(3) group upregulated endothelial nitric oxide synthase and attenuated Evans Blue extravasation at day 3 after LBCCA (P<0.05). Edaravone markedly suppressed accumulation of 4-hydroxy-2-nonenal-modified protein and 8-hydroxy-deoxyguanosine (P<0.01), and loss of oligodendrocytes (P<0.05) in the cerebral white matter at days 3, 7, 14, 21 and 28 after LBCCA. These results were more evident in the ED(3) group. Moreover, at day 21 after LBCCA, spatial memory but not motor function, and axonal damage were significantly improved by three-time treatment of edaravone (P<0.05). Our results indicated that 3-day treatment with edaravone provides protection against WML through endothelial protection and free radical scavenging and suggested that edaravone is potentially useful for the treatment of cognitive impairment.


Subject(s)
Antipyrine/analogs & derivatives , Brain Ischemia/drug therapy , Brain/drug effects , Cerebrovascular Circulation/drug effects , Endothelium, Vascular/drug effects , Free Radical Scavengers/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Antipyrine/pharmacology , Antipyrine/therapeutic use , Apoptosis/drug effects , Axons/drug effects , Axons/pathology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Proliferation/drug effects , Chronic Disease , DNA Damage/drug effects , Edaravone , Endothelium, Vascular/pathology , Free Radical Scavengers/pharmacology , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Memory/drug effects , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type III/biosynthesis , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oxidation-Reduction , Rats , Rats, Wistar
12.
Neuroscience ; 158(2): 665-72, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19032975

ABSTRACT

Pneumonia is a common complication with the highest attributable proportion of deaths in patients with stroke. Cilostazol is a potent type III phosphodiesterase inhibitor, approved as an anti-platelet aggregation agent. The present study was designed to determine the protective mechanism of cilostazol against post-stroke pneumonia using a rat chronic cerebral hypoperfusion model. Rats were subjected to bilateral common carotid artery ligation (LBCCA) and divided randomly into the vehicle group (n=72) and cilostazol group (n=72). Rats of each group were sacrificed at baseline and at days 14, 28 and 42 after LBCCA. Cilostazol significantly improved the swallowing reflex by shortening the latency to elicited swallowing and increasing the numbers of swallows (P<0.05) at 14 days of hypoperfusion. It also decreased the numbers of bacterial colonies grown in cultures from homogenized lungs. Cilostazol markedly upregulated cyclic AMP responsive element binding protein (CREB) phosphorylation, increased tyrosine hydroxylase (TH) expression in the substantial nigra, and maintained dopamine (84.7+/-2.3 vs. 79.2+/-4.1% control; P=0.0512) and substance P levels (86.6+/-7.9 vs. 73.9+/-6.5% control; P<0.05) in the striatum, compared with the vehicle group. Our results indicate that cilostazol improves the swallowing reflex by enhancing the expression of TH through the CREB phosphorylation signaling pathway, and suggest that cilostazol could be useful in preventing pneumonia in the chronic stage of stroke.


Subject(s)
Pneumonia/enzymology , Pneumonia/prevention & control , Tyrosine 3-Monooxygenase/metabolism , Animals , Bacteria/drug effects , Carotid Stenosis/complications , Carotid Stenosis/etiology , Chronic Disease , Cilostazol , Corpus Striatum/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Deglutition/physiology , Disease Models, Animal , Dopamine/metabolism , Enzyme Activation/drug effects , Fibrinolytic Agents/pharmacology , Gene Expression Regulation/drug effects , Ligation/methods , Male , Pneumonia/etiology , Pneumonia/pathology , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Substance P/metabolism , Substantia Nigra/metabolism , Tetrazoles/pharmacology , Time Factors
13.
J Neuroendocrinol ; 20(2): 207-19, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18047553

ABSTRACT

Salusin-alpha and -beta were recently discovered as bioactive endogenous peptides. In the present study, we investigated the effects of chronic osmotic stimuli on salusin-beta-like immunoreactivity (LI) in the rat hypothalamo-neurohypophyseal system. We examined the effects of salusin-beta on synaptic inputs to the rat magnocellular neurosecretory cells (MNCs) of the supraoptic nucleus (SON) and neurohypophyseal hormone release from both freshly dissociated SONs and neurohypophyses in rats. Immunohistochemical studies revealed that salusin-beta-LI neurones and fibres were markedly increased in the SON and the magnocellular division of the paraventricular nucleus after chronic osmotic stimuli resulting from salt loading for 5 days and dehydration for 3 days. Salusin-beta-LI fibres and varicosities in the internal zone of the median eminence and the neurohypophysis were also increased after osmotic stimuli. Whole-cell patch-clamp recordings from rat SON slice preparations showed that salusin-beta did not cause significant changes in the excitatory and inhibitory postsynaptic currents of the MNCs. In vitro hormone release studies showed that salusin-beta evoked both arginine vasopressin (AVP) and oxytocin release from the neurohypophysis, but not the SON. In our hands, in the neurohypophysis, a significant release of AVP and oxytocin was observed only at concentrations from 100 nm and above of salusin-beta. Low concentrations below 100 nm were ineffective both on AVP and oxytocin release. We also measured intracellular calcium ([Ca(2+)](i)) increase induced by salusin-beta on freshly-isolated single nerve terminals from the neurohypophysis devoid of pars intermedia. Furthermore, this salusin-beta-induced [Ca(2+)](i) increase was blocked in the presence of high voltage activated Ca(2+)channel blockers. Our results suggest that salusin-beta may be involved in the regulation of body fluid balance by stimulating neurohypophyseal hormone release from nerve endings by an autocrine/paracrine mechanism.


Subject(s)
Calcium/metabolism , Hypothalamo-Hypophyseal System/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Pituitary Gland, Posterior/metabolism , Pituitary Hormones, Posterior/metabolism , Sodium Chloride/pharmacology , Animals , Arginine Vasopressin/metabolism , Excitatory Postsynaptic Potentials/drug effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Immunohistochemistry , Inhibitory Postsynaptic Potentials/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Intracellular Membranes/metabolism , Male , Osmolar Concentration , Osmosis , Oxytocin/metabolism , Patch-Clamp Techniques , Pituitary Gland, Posterior/drug effects , Pituitary Gland, Posterior/physiology , Presynaptic Terminals/metabolism , Rats , Rats, Wistar , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism
14.
Neuroscience ; 147(1): 71-9, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17512670

ABSTRACT

Accumulation of mutant ubiquitin-B (UBB(+1)) in neurons is considered the hallmark of proteasomal dysfunction in neurodegenerative disorders, however no such evidence in ischemic brain has been reported. We investigated the contribution of UBB(+1) in delayed neuronal death after transient global ischemia. Transient global ischemia was achieved by occlusion of bilateral common carotid arteries for 5 min and reperfusion in male Mongolian gerbils (n=6 per each time point). In the CA1 region, UBB(+1) immunoreactivity appeared in the cytoplasm of pyramidal cells at 30 min post-ischemia, and the density of these neurons increased at day 2 (P<0.001) and further increased at day 4 post-ischemia. Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL)-positive (apoptotic) cells appeared selectively in the CA1 region at day 3 and their density increased further at day 4 post-ischemia (P<0.001). In contrast, UBB(+1) immunoreactivity was only transiently detected from 30 min to 1 day post-ischemia in CA3, dentate gyrus, and frontal cortex, but disappeared at day 2 post-ischemia. No TUNEL-positive cells were observed in these three regions. UBB(+1) mRNA was detected by reverse transcription-polymerase chain reaction in every region of the hippocampus and frontal cortex of ischemic gerbils and even in the non-ischemic control animals, and its expression level was independent of brain region and time after ischemia. Our results indicate induction and selective accumulation of UBB(+1) protein in dying neurons of the CA1 region and suggest that UBB(+1) expression may be induced by proteasomal dysfunction after transient global ischemia.


Subject(s)
Brain Ischemia/metabolism , Hippocampus/metabolism , Pyramidal Cells/metabolism , Ubiquitin/metabolism , Animals , Brain Ischemia/physiopathology , Cell Death/physiology , Frontal Lobe/cytology , Frontal Lobe/metabolism , Gerbillinae , Hippocampus/cytology , Immunohistochemistry , Male , Sequence Deletion/physiology , Ubiquitin-Protein Ligase Complexes/metabolism
15.
Neurology ; 68(7): 528-31, 2007 Feb 13.
Article in English | MEDLINE | ID: mdl-17296920

ABSTRACT

We analyzed the frequency of cardiac embolic sources in 62 patients with acute lacunar stroke vs 50 controls. In post hoc analysis, 11 patients with lacunar stroke having neither hypertension nor diabetes mellitus (non-HDM group) had a higher frequency of right-to-left shunt (RLS) (82%) than patients with risk factors for lacunar stroke. RLS was also independently associated with the non-HDM group. Thus, RLS may contribute to lacunar stroke in patients without risk factors for lacunar stroke.


Subject(s)
Coronary Circulation , Stroke/physiopathology , Aged , Diabetes Complications , Echocardiography, Transesophageal , Female , Humans , Hypertension/complications , Logistic Models , Male , Middle Aged , Prospective Studies , Risk Factors , Stroke/complications , Stroke/diagnostic imaging , Stroke/etiology , Venous Thrombosis/complications
16.
Neuroscience ; 145(2): 520-9, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17241746

ABSTRACT

Hematopoietic prostaglandin D synthase is a key enzyme in synthesis of prostaglandin D. Hematopoietic prostaglandin D synthase is expressed in microglia of the developing mouse brain. This study determined the serial changes and cellular localization of hematopoietic prostaglandin D synthase, and its role in cerebral ischemia/reperfusion injury using C57BL/6 mice (n=84) and bone marrow chimera mice (n=16). The latter mice were selected based on their expression of enhanced green fluorescent protein in bone marrow/blood-derived monocytes/macrophages. The middle cerebral artery was occluded for 60 min, followed by reperfusion. Hematopoietic prostaglandin D synthase expression was examined by immunohistochemistry and Western blotting. Hematopoietic prostaglandin D synthase-positive cells were mainly expressed in the peri-ischemic area at 12 h (P<0.05) and 24 h (P<0.001) after reperfusion, while they were mostly found in the transition area at 48-72 h postreperfusion (P<0.001). There was a significant increase in staining intensity as well as number of hematopoietic prostaglandin D synthase-positive cells in the ischemic core at 5-7 (P<0.001) days postreperfusion. Hematopoietic prostaglandin D synthase-positive cells also co-expressed ionized calcium-binding adapter molecule 1, a marker of microglia/macrophages, and cyclooxygenase-2, but not markers of neurons, oligodendrocytes and astrocytes. Until 72 h postreperfusion, many enhanced green fluorescent protein-positive cells were negative for hematopoietic prostaglandin D synthase, but the number of hematopoietic prostaglandin D synthase-enhanced green fluorescent protein coexpressing cells increased significantly at 5-7 days after reperfusion. Our results indicate that hematopoietic prostaglandin D synthase is mainly produced by endogenous microglia until 72 h after reperfusion, but at 7 days after reperfusion, it is also produced by migrating bone marrow/blood-derived macrophages in the ischemic brain tissue. We speculate that hematopoietic prostaglandin D synthase in the brain has different functions during early and late phases of ischemia.


Subject(s)
Brain Ischemia/enzymology , Brain/enzymology , Intramolecular Oxidoreductases/metabolism , Macrophages/enzymology , Microglia/enzymology , Reperfusion Injury/enzymology , Animals , Brain/blood supply , Brain/physiopathology , Brain Ischemia/physiopathology , Calcium-Binding Proteins/metabolism , Cell Count , Cell Movement/physiology , Cell Proliferation , Cyclooxygenase 2/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoiesis/physiology , Lipocalins , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Prostaglandin D2/biosynthesis , Reperfusion Injury/physiopathology , Transplantation Chimera , Up-Regulation
18.
Neuroscience ; 117(3): 531-9, 2003.
Article in English | MEDLINE | ID: mdl-12617960

ABSTRACT

Brain ischemia induces a marked response of resident microglia and hematopoietic cells including monocytes/macrophages. The present study was designed to assess the distribution of microglia/macrophages in cerebral ischemia using bone marrow chimera mice known to express enhanced green fluorescent protein (EGFP). At 24 h after middle cerebral artery occlusion (MCAO), many round-shaped EGFP-positive cells migrated to the ischemic core and peri-infarct area. At 48-72 h after MCAO, irregular round- or oval-shaped EGFP/ionized calcium-binding adapter molecule 1 (Iba 1)-positive cells increased in the transition zone, while many amoeboid-shaped or large-cell-body EGFP/Iba 1-positive cells were increased in number in the innermost area of ischemia. At 7 days after MCAO, many process-bearing ramified shaped EGFP/Iba 1-positive cells were detected in the transition to the peri-infarct area, while phagocytic cells were distributed in the transition to the core area of the infarction. The distribution of these morphologically variable EGFP/Iba 1-positive cells was similar up to 14 days from MCAO. The present study directly showed the migration and distribution of bone marrow-derived monocytes/macrophages and the relationship between resident microglia and infiltrated hematogenous element in ischemic mouse brain. It is important to study the distribution of intrinsic and extrinsic microglia/macrophage in ischemic brain, since such findings may allow the design of appropriate gene-delivery system using exogenous microglia/macrophages to the ischemic brain area.


Subject(s)
Bone Marrow/pathology , Brain Ischemia/pathology , Cell Movement , Microglia/pathology , Animals , Bone Marrow/radiation effects , Brain Ischemia/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/radiation effects , Cell Count , Cell Movement/radiation effects , Central Nervous System/injuries , Central Nervous System/physiopathology , Central Nervous System/radiation effects , Chimera/metabolism , Dose-Response Relationship, Radiation , Fluorouracil/toxicity , Green Fluorescent Proteins , Immunohistochemistry/methods , Immunosuppressive Agents , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Luminescent Proteins/metabolism , Macrophages/pathology , Macrophages/radiation effects , Mice , Mice, Transgenic , Microfilament Proteins , Microglia/radiation effects , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/radiation effects , Time Factors , Transplants , Whole-Body Irradiation
19.
J Neurochem ; 82(4): 953-60, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12358801

ABSTRACT

Neuronal progenitor cells (NPC) are particularly suited as the target population for genetic and cellular therapy of neurological disorders such as Parkinson's disease or stroke. However, genetic modification of these cells using retroviral vectors remains a great challenge because of the low transduction rate and the need for fetal calf serum (FCS) during the transduction process that induces the cell differentiation to mature neurons. To overcome these problems, we developed a new retrovirus production system in which the simplified retroviral vector GCDNsap engineered to be resistant to denovo methylation was packaged in the vesicular stomatitis virus G protein (VSV-G), concentrated by centrifugation, and resuspended in serum-free medium (StemPro-34 SFM). In transduction experiments using enhanced green fluorescent protein (EGFP) as a marker, the concentrated FCS-free virus supernatant infected NPC at a high rate, while maintaining the ability of these cells to self-renew and differentiate in vitro. When such cells were grafted into mouse brains, EGFP-expressing NPC were detected in the region around the injection site at 8 weeks post transplantation. These findings suggest that the gene transfer system described here may provide a useful tool to genetically modify NPC for treatments of neurological disorders.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/pharmacology , Membrane Glycoproteins , Nervous System Diseases/therapy , Retroviridae/genetics , Viral Envelope Proteins/genetics , Animals , Cells, Cultured , Clone Cells/virology , Corpus Striatum/cytology , Feasibility Studies , Genes, Reporter , Genetic Vectors/genetics , Graft Survival , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/virology , Transduction, Genetic , Viral Envelope Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 98(19): 10918-23, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11535810

ABSTRACT

Adeno-associated virus (AAV) vector delivery of an Apaf-1-dominant negative inhibitor was tested for its antiapoptotic effect on degenerating nigrostriatal neurons in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease. The wild-type caspase recruitment domain of Apaf-1 was used as a dominant negative inhibitor of Apaf-1 (rAAV-Apaf-1-DN-EGFP). An AAV virus vector was used to deliver it into the striatum of C57 black mice, and the animals were treated with MPTP. The number of tyrosine hydroxylase-positive neurons in the substantia nigra was not changed on the rAAV-Apaf-1-DN-EGFP injected side compared with the noninjected side. We also examined the effect of a caspase 1 C285G mutant as a dominant negative inhibitor of caspase 1 (rAAV-caspase-1-DN-EGFP) in the same model. However, there was no difference in the number of tyrosine hydroxylase-positive neurons between the rAAV-caspase-1-DN-EGFP injected side and the noninjected side. These results indicate that delivery of Apaf-1-DN by using an AAV vector system can prevent nigrostriatal degeneration in MPTP mice, suggesting that it could be a promising therapeutic strategy for patients with Parkinson's disease. The major mechanism of dopaminergic neuronal death triggered by MPTP seems to be the mitochondrial apoptotic pathway.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Apoptosis , Dopamine Agents/metabolism , Parkinson Disease/drug therapy , Proteins/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Apoptotic Protease-Activating Factor 1 , Biological Transport , Caspase 1/metabolism , Corpus Striatum/metabolism , Dependovirus , Dopamine Agents/pharmacology , Gene Expression , Genetic Vectors , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Parkinson Disease/metabolism , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...