Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Neuroscience ; 121(3): 605-17, 2003.
Article in English | MEDLINE | ID: mdl-14568021

ABSTRACT

Multiple drug resistance occurs when cells fail to respond to chemotherapy. Although it has been established that the drug efflux protein P-glycoprotein protects the brain from xenobiotics, the mechanisms involved in the regulation of expression of multiple drug resistance genes and proteins are not fully understood. Re-entry into the cell cycle and integrity of the p53 signaling pathway have been proposed as triggers of multiple drug resistance expression in tumor cells. Whether this regulation occurs in non-tumor CNS tissue is not known. Since multiple drug resistance overexpression has been reported in glia and blood vessels from epileptic brain, we investigated the level of expression of multidrug resistance protein, multidrug resistance-associated proteins and lung resistance protein in endothelial cells and astrocytes isolated from epileptic patients or studied in situ in surgical tissue samples by double label immunocytochemistry. Reverse transcriptase-polymerase chain reaction and Western blot analyses revealed that multiple drug resistance, multidrug resistance protein, and lung resistance protein are expressed in these cells. Given that lung resistance proteins have been reported to be preferentially expressed by tumors, we investigated expression of tumor suppressor genes in epileptic cortices. The pro-apoptotic proteins p53 and p21 could not be detected in "epileptic" astrocytes, while endothelial cells from the same samples readily expressed these proteins, as did normal brain astroglia and normal endothelial cells. Other apoptotic markers were also absent in epileptic glia. Our results suggest a possible link between loss of p53 function and expression of multiple drug resistance in non-tumor CNS cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Astrocytes/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Astrocytoma/metabolism , Brain/anatomy & histology , Brain/pathology , Brain Neoplasms/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chemokines, CC/metabolism , Endothelium/metabolism , Epilepsy/metabolism , Female , Gene Expression , Humans , Immunoblotting/methods , Immunohistochemistry/methods , In Situ Hybridization , Indoles/metabolism , Infant , Male , Microscopy, Confocal , Middle Aged , Neoplasm Proteins/metabolism , RNA/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Vault Ribonucleoprotein Particles/metabolism
2.
J Cereb Blood Flow Metab ; 21(12): 1430-5, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11740204

ABSTRACT

The authors previously reported that mRNA for macrophage inflammatory protein-1alpha (MIP-1 alpha), a member of the CC chemokines, was expressed in glial cells after focal cerebral ischemia in rats. However, the function of chemokines in the ischemic brain remains unclear. Recently, viral macrophage inflammatory protein-II (vMIP-II), a chemokine analogue encoded by human herpesvirus-8 DNA, has been demonstrated to have antagonistic activity at several chemokine receptors. In the present study, the effects of vMIP-II and MIP-1alpha on ischemic brain injury were examined in mice to elucidate the roles of chemokines endogenously produced in the ischemic brain. Intracerebroventricular injection of vMIP-II (0.01-1 microg) reduced infarct volume in a dose-dependent manner when examined 48 hours after 1-hour middle cerebral artery occlusion followed by reperfusion. However, 1 microg MIP-1alpha increased infarct volume in the cortical region. These results supported the possibility that chemokines endogenously produced in the brain are involved in ischemic injury, and that chemokine receptors are potential targets for therapeutic intervention of stroke.


Subject(s)
Brain Ischemia/drug therapy , Chemokines, CC/pharmacology , Chemokines/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Animals , Brain Infarction/drug therapy , Brain Infarction/metabolism , Brain Ischemia/metabolism , Cerebrovascular Circulation/drug effects , Chemokine CCL3 , Chemokine CCL4 , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Injections, Intraventricular , Macrophage Inflammatory Proteins/metabolism , Male , Mice
3.
Neuroreport ; 12(13): 2947-50, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11588608

ABSTRACT

PEPA (4-[2-(Phenylsulphonylamino)ethylthio]-2,6-difluorophenoxyacetamide) is a recently developed allosteric potentiator of AMPA receptors that preferentially affects flop splice variants. We tested the effects of PEPA on ischemia-induced memory deficit in rats. Permanent unilateral occlusion of the middle cerebral artery induced severe impairment of performance of rats in the Morris water maze test. Repeated intravenous administration of PEPA (1, 3, 10 mg/kg/day for 10 days) improved test performance. In contrast, a corresponding dose of aniracetam, a representative potentiator of AMPA receptor, did not significantly improve test performance. Thus, PEPA is more effective than aniracetam in reversing impaired memory function as assessed by the Morris water maze test; and PEPA may be an effective compound for the treatment of impaired memory.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Excitatory Amino Acid Agonists/pharmacology , Memory Disorders/drug therapy , Nootropic Agents/pharmacology , Phenoxyacetates/pharmacology , Receptors, AMPA/drug effects , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Brain/metabolism , Brain/physiopathology , Brain Ischemia/complications , Brain Ischemia/physiopathology , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/physiopathology , Pyrrolidinones/pharmacology , Rats , Rats, Inbred SHR , Receptors, AMPA/metabolism
4.
Stroke ; 32(8): 1906-11, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11486124

ABSTRACT

BACKGROUND AND PURPOSE: The seleno-organic compound ebselen has both antioxidant and anti-inflammatory properties. Although ebselen has been shown to protect the brain against stroke, it is unclear how ebselen provides neuroprotection. In the present study the authors examined whether ebselen inhibits neuronal apoptosis resulting from transient focal cerebral ischemia in mice. The cytochrome c release and DNA fragmentation, both of which are biochemical markers of apoptosis, were compared between vehicle- and ebselen-treated mice. METHODS: Cerebral ischemia was induced by transient middle cerebral artery occlusion for 30 minutes in ICR mice under halothane anesthesia. Ebselen (10 mg/kg) was given orally twice, 30 minutes before ischemia and 12 hours after reperfusion. By Western blot analysis, we examined release of mitochondrial cytochrome c. To evaluate brain damage, the brain sections were treated for terminal deoxynucleotidyl transferase-mediated DNA nick-end labeling (TUNEL) and Nissl staining. Prolonged neuroprotective efficacy of ebselen was determined by counting neuronal nuclei (NeuN) immunopositive cells at 21 days after ischemia. RESULTS: - Cytochrome c release was detected in the ischemic hemisphere at 3 to 24 hours after ischemia. Ebselen treatment diminished the cytochrome c release at 12 and 24 hours. In addition, ebselen decreased both DNA fragmentation determined by TUNEL and brain damage volume at 3 days after ischemia. Furthermore, ebselen increased the number of NeuN immunopositive cells at 21 days after ischemia. CONCLUSIONS: These results indicate that ebselen attenuates ischemic neuronal apoptosis by inhibiting cytochrome c release. Ebselen may be a potential compound in stroke therapy.


Subject(s)
Azoles/pharmacology , Cytochrome c Group/metabolism , DNA Fragmentation/drug effects , Ischemic Attack, Transient/drug therapy , Mitochondria/drug effects , Organoselenium Compounds/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , In Situ Nick-End Labeling , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Isoindoles , Male , Mice , Mice, Inbred ICR , Mitochondria/enzymology , Mitochondria/pathology , Neuroprotective Agents/pharmacology
5.
Proc Natl Acad Sci U S A ; 98(20): 11569-74, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11504919

ABSTRACT

Brain subjected to acute ischemic attack caused by an arterial blockage needs immediate arterial recanalization. However, restoration of cerebral blood flow can cause tissue injury, which is termed reperfusion injury. It is important to inhibit reperfusion injury to achieve greater brain protection. Because oxidative stress has been shown to activate mitogen-activated protein kinases (MAPKs), and because oxidative stress contributes to reperfusion injury, MAPK may be a potential target to inhibit reperfusion injury after brain ischemia. Here, we demonstrate that reperfusion after forebrain ischemia dramatically increases phosphorylation level of extracellular signal-regulated kinase 2 (ERK2) in the gerbil hippocampus. In addition, i.v. administration of U0126 (100-200 mg/kg), a specific inhibitor of MEK (MAPK/ERK kinase), protects the hippocampus against forebrain ischemia. Moreover, treatment with U0126 at 3 h after ischemia significantly reduces infarct volume after transient (3 h) focal cerebral ischemia in mice. This protection is accompanied by reduced phosphorylation level of ERK2, substrates for MEK, in the damaged brain areas. Furthermore, U0126 protects mouse primary cultured cortical neurons against oxygen deprivation for 9 h as well as nitric oxide toxicity. These results provide further evidence for the role of MEK/ERK activation in brain injury resulting from ischemia/reperfusion, and indicate that MEK inhibition may increase the resistance of tissue to ischemic injury.


Subject(s)
Butadienes/therapeutic use , Enzyme Inhibitors/therapeutic use , Ischemic Attack, Transient/prevention & control , MAP Kinase Kinase Kinase 1 , Nitriles/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Gerbillinae , In Vitro Techniques , Male , Mice , Mice, Inbred ICR , Reperfusion , Time Factors
6.
Neurosci Lett ; 305(1): 41-4, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11356303

ABSTRACT

Matrix metalloproteinases (MMPs) are proteolytic enzymes that can degrade the extracellular matrix. MMP-9 and MMP-2 have been implicated in brain injury formation. The authors examined the effect of MMP inhibitor KB-R7785 on brain infarct formation resulting from permanent focal cerebral ischemia in mice. Ischemia was induced by intraluminal middle cerebral artery occlusion (MCAO) in mice under halothane anesthesia. Zymography was conducted to measure the MMPs activity in ischemic brain tissues. Injection of KB-R7785 (100 mg/kg) 30 min before MCAO significantly decreased both MMP-9 activity and infarct volume determined at 24 h. In addition, KB-R7785 injected twice at 1 and 4.5 h after MCAO significantly decreased infarct volume. These results indicate that KB-R7785 has a protective efficacy against focal cerebral ischemia, and our data provide further evidence that MMP-9 contributes to brain infarct formation.


Subject(s)
Brain Ischemia/pathology , Brain/drug effects , Brain/pathology , Glycine/pharmacology , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/pharmacology , Animals , Brain/enzymology , Brain Ischemia/enzymology , Cerebral Infarction/enzymology , Cerebral Infarction/pathology , Glycine/analogs & derivatives , Male , Mice , Mice, Inbred ICR
7.
J Neurochem ; 77(1): 50-62, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11279261

ABSTRACT

Previously we found that some cyclopentenone prostaglandin derivatives promoted neurite outgrowth from PC12 cells and dorsal root ganglia explants in the presence of nerve growth factor; and so we referred to them as neurite outgrowth-promoting prostaglandins (NEPPs). In this study, NEPPs protected HT22 cells against oxidative glutamate toxicity. NEPP6, one of the most effective promoters of neurite outgrowth in PC12 cells, protected the cells most potently among NEPPs 1--10. Several derivatives, NEPPs 11--19, were newly synthesized based on the chemical structure of NEPP6. NEPP11 had a more potent neuroprotective effect than NEPP6. NEPP11 also prevented the death of cortical neurons induced by various stimuli and reduced ischemic brain damage in mice. Biotinylated compounds of NEPPs were synthesized to investigate their cellular accumulation. NEPP6-biotin protected the cells and emitted potent signals from the cells. In contrast, biotinylated non-neuroprotective derivatives emitted much weaker signals. These results suggest that NEPPs are novel types of neurotrophic compounds characterized by their dual biological activities of promoting neurite outgrowth and preventing neuronal death and that their accumulation in the cells is closely associated with their neuroprotective actions.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Prostaglandins/pharmacology , Animals , Biotin/analogs & derivatives , Biotin/chemistry , Biotin/pharmacology , Brain Ischemia/drug therapy , Cell Survival/drug effects , Cells, Cultured , Cyclopentanes/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Mice , Microinjections , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/chemistry , Neurites/drug effects , Neurons/cytology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Prostaglandin D2/analogs & derivatives , Prostaglandins/administration & dosage , Prostaglandins/chemistry , Structure-Activity Relationship
8.
J Cereb Blood Flow Metab ; 20(9): 1301-5, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10994851

ABSTRACT

The protein kinase Akt/PKB has been implicated in antiapoptosis and neuronal survival. The authors now show that Akt is phosphorylated in the hippocampus during the early reperfusion period after 3.5 minutes bilateral carotid artery occlusion (BCAO) in the gerbil. Repeated sublethal ischemia induces ischemic tolerance, which is known as ischemic preconditioning. Ischemic preconditioning does not affect the amount of Akt protein, but rather decreases the phosphorylation of Akt at Ser-473 after 10 minutes reperfusion after 3.5 minutes BCAO. These results suggest that although Akt may play a role in neuronal survival after ischemia, it may not play a role in ischemic tolerance by preconditioning.


Subject(s)
Brain Ischemia , Hippocampus/blood supply , Proto-Oncogene Proteins/physiology , Animals , Cell Death , Gerbillinae , Hippocampus/physiopathology , Ischemic Preconditioning , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt
9.
Neurosci Lett ; 288(2): 163-6, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10876086

ABSTRACT

Oxidative stress is implicated in the pathogenesis of neuronal degenerative diseases. Oxidative stress has been shown to activate extracellular signal-regulated kinases (ERK)1/2. We investigated the role of these mitogen-activated protein kinases (MAPKs) in oxidative neuronal injury by using a mouse hippocampal cell line (HT22) and rat primary cortical cultures. Here, we show that a novel MAPK/ERK kinase (MEK) specific inhibitor U0126 profoundly protected HT22 cells against oxidative stress induced by glutamate, which was accompanied by an inhibition of phosphorylation of ERK1/2. U0126 also protected rat primary cultured cortical neurons against glutamate or hypoxia. However, U0126 was not protective against death caused by tumor necrosis factor alpha (TNFalpha), A23187, or staurosporine. These results indicate that MEK plays a central role in the neuronal death caused by oxidative stress.


Subject(s)
Butadienes/pharmacology , Cerebral Cortex/enzymology , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/enzymology , Neuroprotective Agents/pharmacology , Nitriles/pharmacology , Oxidative Stress/drug effects , Animals , Cell Line , Mice , Mitogen-Activated Protein Kinases/physiology , Neurons/drug effects , Neurons/physiology , Oxidative Stress/physiology , Rats , Rats, Wistar
10.
J Cell Biol ; 149(3): 613-22, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10791975

ABSTRACT

Caspase-11, a member of the murine caspase family, has been shown to be an upstream activator of caspase-1 in regulating cytokine maturation. We demonstrate here that in addition to its defect in cytokine maturation, caspase-11-deficient mice have a reduced number of apoptotic cells and a defect in caspase-3 activation after middle cerebral artery occlusion (MCAO), a mouse model of stroke. Recombinant procaspase-11 can autoprocess itself in vitro. Purified active recombinant caspase-11 cleaves and activates procaspase-3 very efficiently. Using a positional scanning combinatorial library method, we found that the optimal cleavage site of caspase-11 was (I/L/V/P)EHD, similar to that of upstream caspases such as caspase-8 and -9. Our results suggest that caspase-11 is a critical initiator caspase responsible for the activation of caspase-3, as well as caspase-1 under certain pathological conditions.


Subject(s)
Caspase 1/metabolism , Caspases/metabolism , Animals , Apoptosis , Brain Ischemia/enzymology , Brain Ischemia/pathology , Caspase 3 , Caspase Inhibitors , Caspases/deficiency , Caspases/genetics , Caspases, Initiator , Cell Line , Combinatorial Chemistry Techniques , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Immunohistochemistry , Kinetics , Mice , Mice, Knockout , Protein Precursors/metabolism , Recombinant Proteins , Stroke/enzymology , Substrate Specificity
11.
J Neurosci ; 20(9): 3175-81, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10777781

ABSTRACT

DNA methylation is important for controlling the profile of gene expression and is catalyzed by DNA methyltransferase (MTase), an enzyme that is abundant in brain. Because significant DNA damage and alterations in gene expression develop as a consequence of cerebral ischemia, we measured MTase activity in vitro and DNA methylation in vivo after mild focal brain ischemia. After 30 min middle cerebral artery occlusion (MCAo) and reperfusion, MTase catalytic activity and the 190 kDa band on immunoblot did not change over time. However, [(3)H]methyl-group incorporation into DNA increased significantly in wild-type mice after reperfusion, but not in mutant mice heterozygous for a DNA methyltransferase gene deletion (Dnmt(S/+)). Dnmt(S/+) mice were resistant to mild ischemic damage, suggesting that increased DNA methylation is associated with augmented brain injury after MCA occlusion. Consistent with this formulation, treatment with the MTase inhibitor 5-aza-2'-deoxycytidine and the deacetylation inhibitor trichostatin A conferred stroke protection in wild-type mice. In contrast to mild stroke, however, DNA methylation was not enhanced, and reduced dnmt gene expression was not protective in an ischemia model of excitotoxic/necrotic cell death. In conclusion, our results demonstrate that MTase activity contributes to poor tissue outcome after mild ischemic brain injury.


Subject(s)
Brain Ischemia/metabolism , DNA Methylation , Gene Expression/physiology , Methyltransferases/metabolism , Reperfusion Injury/metabolism , Animals , Brain/metabolism , Brain Ischemia/prevention & control , Gene Expression/genetics , Infarction, Middle Cerebral Artery/metabolism , Methyltransferases/genetics , Mice , Mice, Transgenic
12.
No Shinkei Geka ; 27(10): 941-6, 1999 Oct.
Article in Japanese | MEDLINE | ID: mdl-10535084

ABSTRACT

It is difficult to treat ruptured aneurysms with symptomatic vasospasm. Although direct surgery for such cases is associated with poor outcomes, conservative therapy has the risk of both rerupture and infarction. In two cases of ruptured aneurysms with symptomatic vasospasm, we performed aneurysmal coil embolization with Guglielmi electrodetatchable coils (GDC). At the same time we performed percutaneous transluminal angioplasty (PTA) with papaverine infusion. In both cases, rerupture did not occur and PTA was effective angiographically. A good outcome was achieved in case 1. However, broad cerebral infarction occurred in case 2, in which the patient had shown severe symptomatic vasospasm on admission. In advanced cases, such as in case 2, the outcome is poor. The aneurysm may not be able to be approached before PTA because of severe vasospasm. In such cases, PTA must be performed carefully to avoid aneurysmal rerupture. Intraarterial papaverine infusion is safer than PTA for severe spasm in distal vessels. However the efficacy of papaverine is known to be transient in many cases. It is often difficult to determine the exact relationship between branches and the aneurysm in the presence of vasospasm. In such cases, we recommend that the rupture point be packed and that the aneurysmal neck remain unpacked. After vasospasm is cured and good general condition has been recovered, direct surgery can be performed. In summary, endovascular surgery is an effective option for treatment of ruptured aneurysm with symptomatic vasospasm.


Subject(s)
Aneurysm, Ruptured/therapy , Angioplasty, Balloon , Embolization, Therapeutic , Intracranial Aneurysm/therapy , Vasospasm, Intracranial/complications , Aged , Aneurysm, Ruptured/complications , Aneurysm, Ruptured/diagnostic imaging , Female , Humans , Intracranial Aneurysm/complications , Intracranial Aneurysm/diagnostic imaging , Middle Aged , Papaverine/administration & dosage , Radiography , Vasospasm, Intracranial/diagnostic imaging
13.
Proc Natl Acad Sci U S A ; 96(22): 12866-9, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10536014

ABSTRACT

The MEK1 (MAP kinase/ERK kinase)/ERK (extracellular-signal-responsive kinase) pathway has been implicated in cell growth and differentiation [Seger, R. & Krebs, E. G. (1995) FASEB J. 9, 726-735]. Here we show that the MEK/ERK pathway is activated during focal cerebral ischemia and may play a role in inducing damage. Treatment of mice 30 min before ischemia with the MEK1-specific inhibitor PD98059 [Alessi, D. R., Cuenda, A., Cohen, P. , Dudley, D. T. & Saltiel, A. R. (1995) J. Biol. Chem. 270, 27489-27494] reduces focal infarct volume at 22 hr after ischemia by 55% after transient occlusion of the middle cerebral artery. This is accompanied by a reduction in phospho-ERK1/2 immunohistochemical staining. MEK1 inhibition also results in reduced brain damage 72 hr after ischemia, with focal infarct volume reduced by 36%. This study indicates that the MEK1/ERK pathway contributes to brain injury during focal cerebral ischemia and that PD98059, a MEK1-specific antagonist, is a potent neuroprotective agent.


Subject(s)
Brain Ischemia/drug therapy , Enzyme Inhibitors/therapeutic use , Flavonoids/therapeutic use , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases , Animals , Body Temperature Regulation/drug effects , Brain Ischemia/enzymology , Brain Ischemia/pathology , Cardiovascular System/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Imidazoles/pharmacology , Immunohistochemistry , MAP Kinase Kinase 1 , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphorylation , Pyridines/pharmacology , Respiration/drug effects , p38 Mitogen-Activated Protein Kinases
14.
J Oral Sci ; 41(1): 5-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10230154

ABSTRACT

This study was undertaken to develop a three-dimensional reconstruction system using magnetic resonance (MR) images in order to visualize three-dimensional images of the temporomandibular joint (TMJ) including the disk. The computerized reconstruction program (written using Visual Basic for Windows, Microsoft Corp.) could reliably generate three-dimensional images of the TMJ. Image processing techniques made the tracing of images unnecessary, reduced complex human manipulation and associated measurement errors. This system, capable of treating fifty thousand pixels or more, generates smooth three-dimensional images of the TMJ.


Subject(s)
Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Temporomandibular Joint/pathology , Data Display , Humans , Image Processing, Computer-Assisted/instrumentation , Magnetic Resonance Imaging/instrumentation , Mandibular Condyle/pathology , Reproducibility of Results , Software , Temporomandibular Joint Disc/pathology , Temporomandibular Joint Disorders/pathology
15.
J Dermatol Sci ; 19(1): 31-6, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9890372

ABSTRACT

It is well known that Staphylococcus aureus (S. aureus) proliferates on the moist skin lesion of atopic dermatitis. Reduction of bacteria colonization from skin lesions by antibiotics has been reported to be effective for the treatment of atopic dermatitis. S. aureus produces superantigens which can activate T cells and possibly enhance the inflammatory reaction. Photo(chemo)therapy has been successfully used for the treatment of severe cases of atopic dermatitis. We have previously reported that photo(chemo)therapy had bacteriostatic effect on S. aureus. Now we examined the effect of UVB and psoralen plus UVA (PUVA) on superantigen production from S. aureus. We isolated S. aureus from six atopic dermatitis patients. S. aureus was irradiated in vitro with UVB (0, 5, 10 mJ/cm2) or PUVA (0.001% psoralen plus 0, 5, 10 mJ/cm2 UVA) and incubated 4 h with 100 strokes per min. After incubation, the amounts of superantigens in the supernatant were measured using ELISA kit. The production of superantigens decreased in an ultraviolet dose-dependent manner. The suppressive effects of UV radiation on superantigen production may be involved in the therapeutic efficacy of photo(chemo)therapy for atopic dermatitis.


Subject(s)
Staphylococcus aureus/radiation effects , Superantigens/radiation effects , Ultraviolet Rays , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/radiotherapy , Dose-Response Relationship, Radiation , Humans , PUVA Therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Superantigens/biosynthesis , Superantigens/drug effects
16.
Neuroscience ; 94(4): 1213-8, 1999.
Article in English | MEDLINE | ID: mdl-10625061

ABSTRACT

Necrotic and apoptotic cell death both play a role mediating tissue injury following brain trauma. Caspase-1 (interleukin-1beta converting enzyme) is activated and oligonucleosomal DNA fragmentation is detected in traumatized brain tissue. Reduction of tissue injury and free radical production following brain trauma was achieved in a transgenic mouse expressing a dominant negative inhibitor of caspase-1 in the brain. Neuroprotection was also conferred by pharmacological inhibition of caspase-1 by intracerebroventricular administration of the selective inhibitor of caspase-1, acetyl-Tyr-Val-Ala-Asp-chloromethyl-ketone or the non-selective caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone. These results indicate that inhibition of caspase-1-like caspases reduces trauma-mediated brain tissue injury. In addition, we demonstrate an in vivo functional interaction between interleukin-1beta converting enyzme-like caspases and free radical production pathways, implicating free radical production as a downstream mediator of the caspase cell death cascade.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Brain Injuries/pathology , Brain/pathology , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Hydroxyl Radical/metabolism , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Brain Injuries/genetics , Caspase 1/physiology , DNA Fragmentation , Interleukin-1/metabolism , Mice , Mice, Transgenic/genetics
17.
J Cereb Blood Flow Metab ; 18(10): 1071-6, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9778183

ABSTRACT

Apoptotic cell death is prominent in neurodegenerative disorders, such as Alzheimer's disease and Huntington's disease, and is found in cerebral ischemia. Using a murine model of delayed cell death, we determined that cleavage of zDEVD-amino-4-trifluoromethyl coumarin (zDEVD-afc) in brain homogenate, a measure of caspase activation, increased initially 9 hours after brief (30 minutes) middle cerebral artery occlusion along with caspase-3p20 immunoreactive cleavage product as determined by immunoblotting. zDEVD-afc cleavage activity was blocked by pretreatment or posttreatment with the caspase-inhibitor N-benzyloxycarbonyl-Asp(OMe)-Glu(OMe)-Val-Asp(OMe)-fluoromethyl-ketone (zDEVD-fmk), and ischemic damage was reduced when the drug was injected up to 9 hours after reperfusion. The protection was long lasting (21 days). Hence, the period before caspase activation defined the therapeutic opportunity for this neuroprotective agent after mild ischemic brain injury. Prolonged protection after caspase inhibition plus the extended treatment window may be especially relevant to the treatment of neurodegenerative disorders.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/therapy , Caspases/metabolism , Animals , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Caspase Inhibitors , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation/drug effects , Male , Mice , Mice, Inbred Strains , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Time Factors
18.
Neurosurgery ; 43(4): 900-7; discussion 907-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9766318

ABSTRACT

OBJECTIVE: The recent availability of transgenic mice enables us to study the functional role of single gene products in cerebral ischemia. To establish an experimental murine model of transient forebrain ischemia, this study examined the temporal profile of ischemic neuronal damage in the striatum after bilateral common carotid artery occlusion. METHODS: C57BL/6 mice, which are frequently used for genetic manipulations, were subjected to 15-minute bilateral common carotid artery occlusion. Ischemic injury was examined (4, 8, 24, 48, and 96 h after reperfusion) by Nissl staining, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick-end-labeling, and nuclear staining with Hoechst 33258 dye. RESULTS: Regional cerebral blood flow was decreased to 11 +/- 6% of control values during the ischemic insult. Striatal injury was observed in 95% of animals examined after 15-minute bilateral common carotid artery occlusion. The number of small and medium-size neurons in the striatum was significantly (P < 0.05) decreased 8 hours after reperfusion and continued to decrease until 96 hours, whereas the number of large neurons remained constant. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick-end-labeling-positive cells appeared in the dorsomedial region of the striatum 48 hours after the ischemic insult and throughout the striatum 96 hours after the ischemic insult. Brain sections stained with Hoechst 33258 dye also demonstrated apoptotic nuclei 96 hours after the ischemic insult. CONCLUSION: Striatal injury after transient forebrain ischemia is reproducible in C57BL/6 mice and is a good model to study the molecular mechanisms of ischemic injury, including delayed neuronal death, using transgenic mice.


Subject(s)
Brain Ischemia/pathology , Carotid Stenosis/pathology , Corpus Striatum/blood supply , Dominance, Cerebral/physiology , Ischemic Attack, Transient/pathology , Animals , Apoptosis/physiology , Brain Mapping , Corpus Striatum/pathology , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Necrosis , Neurons/pathology , Reperfusion Injury/pathology
19.
J Oral Sci ; 40(2): 89-94, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9680766

ABSTRACT

Kinematic imaging of the temporomandibular joint (TMJ) was applied for diagnosis of TMJ disorders using an ultrasonic diagnostic imaging system. Patients with a normal TMJ (male, 24 y 1 mon) and a symptomatic TMJ (female, 20 y 2 mon) were selected for imaging. The transducer must be placed in a specific location in order to propagate ultrasound through soft tissue because it is difficult for ultrasound to penetrate bone such as the condyle and the eminence. Therefore the ultrasonic images were not taken in sagittal cross-section, as is the case with magnetic resonance images. The ultrasonic diagnostic imaging system showed a transverse cross-section and no hard tissue images. It was difficult to become accustomed to these images, thus making it difficult to find differences between the normal TMJ and the symptomatic TMJ on the basis of static ultrasonic images alone. However a difference between the kinematic images of the normal and symptomatic TMJ was observed during jaw opening. Irregularity in the striated pattern of the soft tissue surrounding the condyle was observed in the image of the symptomatic TMJ. In order to make a precise diagnosis using ultrasonic imaging, it may be useful to understand the kinematics of the soft tissue surrounding the TMJ during jaw opening and closing.


Subject(s)
Temporomandibular Joint Disorders/diagnostic imaging , Temporomandibular Joint/diagnostic imaging , Adult , Cartilage, Articular/diagnostic imaging , Female , Humans , Joint Dislocations/diagnostic imaging , Male , Mandibular Condyle/diagnostic imaging , Range of Motion, Articular , Temporomandibular Joint Disc/diagnostic imaging , Ultrasonography
20.
Neurosci Lett ; 248(1): 41-4, 1998 May 22.
Article in English | MEDLINE | ID: mdl-9665659

ABSTRACT

Poly(ADP-ribose) synthetase (PARS) activation, a downstream event of nitric oxide (NO) neurotoxicity has been implicated in cerebral reperfusion injury. The aim of our study was to identify the trigger of PARS activation during stroke. Formation of poly(ADP-ribose) profoundly increased in the early phase of reperfusion. Poly(ADP-ribose) formation was attenuated in mice deficient for neuronal NO synthase (nNOS). We next tested in glioma cells whether NO, or peroxynitrite (a cytotoxic oxidant formed from NO and superoxide) is the actual trigger of PARS activation. Peroxynitrite, but not various NO donors, activated PARS and suppressed cellular viability in a PARS-dependent fashion. Thus, nNOS is responsible for PARS activation in stroke. PARS activation, however, is not a direct result of NO production, but it occurs via peroxynitrite formation.


Subject(s)
Brain Ischemia/enzymology , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Nitrates/physiology , Nitric Oxide Synthase/physiology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Enzyme Activation , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Nitrates/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Oxidants/physiology , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...