Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Neurobiol ; 56(1): 658-670, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29785469

ABSTRACT

Prolongation of the T2 relaxation time, an increase in T2-weighted signal intensity (T2-SI), and a decrease in the apparent diffusion coefficient (ADC) calculated from diffusion-weighted images (DWI) on magnetic resonance imaging (MRI) are conventional indicators of the vasogenic (interstitial) or cytotoxic (cellular) cerebral edema that develops after ischemic stroke. However, these parameters obtained on stroke imaging have not given us a precise threshold at which we can determine the viability or vulnerability of the tissue, allowing us to decide on an intervention that will help reversible tissue in the acute phase. Here, we introduce a new indicator-the essential diffusion coefficient or EDC, calculated from the T2-SI and ADC-that permits detection of irreversible brain damage after induction of experimental, focal cerebral ischemia. Our three-vessel occlusion (3-VO) method (Yang et al. Eur Neurol 71:4-18, 2014) was applied to investigate early changes on 7-T MRI. In the 3-VO model, which targets only a part of the cortex, animals seldom die at least within 24 h. The T2-SI and the ADC value were monitored, starting at 60 min after reperfusion, and every 30-60 min, for 10 h after the induction of focal ischemia. The region of interest (ROI) was set in each of the following: (1) the ischemic core (the dead zone); (2) the medial border area (the dying/dead mixed zone, including the ischemic penumbra); (3) the lateral border area (the surviving zone after the ischemic stress, where the rCBF is above the threshold for death); and (4) The intact area (outside the ischemic zone). The diagnosis was made by histological analysis performed 24 h after reperfusion. Significant increases in the T2-SI were observed, in ROI-1 at 1 h, in ROI-2 at 2.5 h, and in ROI-3 at 4 h post-reperfusion (1.10, 1.11, or 1.11; > 1.10, respectively, p < 0.001). Significant reductions in the ADC were also observed in ROI-1, ROI-2, and ROI-3, at 1 h post-reperfusion (0.55, 0.52, or 0.58; < 0.60, respectively, p < 0.001), indicating that both types of cerebral edema develop simultaneously in the acute phase. In the EDC analysis, from 5.0 h post-reperfusion, the value in the dying/dead zone (ROI-1 and ROI-2) was consistently reduced to < 50%, showing repeated, significant differences from the value in the surviving zone (ROI-3). A reduction in the EDC to below 50% indicated irreversible tissue damage, with transformation to cerebral infarction. We could detect a sign of cerebral infarction (initial necrosis-like irreversible lesion) as early as 5.25 h after the onset of ischemia. Although the biological time that depends on the body weight must be different between mice and humans, the earliest irreversible tissue damage or tissue destruction (to have achieved the risk of hemorrhagic transformation) that progressed after invisible or silent cell death in the ultra-acute phase, seems to occur at a similar time point.


Subject(s)
Cerebral Infarction/diagnostic imaging , Cerebral Infarction/diagnosis , Magnetic Resonance Imaging , Animals , Brain Edema/pathology , Cerebral Infarction/surgery , Diffusion Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Vital Signs
2.
PLoS One ; 12(7): e0180981, 2017.
Article in English | MEDLINE | ID: mdl-28686706

ABSTRACT

Plasminogen (Plg) is a precursor of plasmin that degrades fibrin. A race-specific A620T mutation in Plg, also known as Plg-Tochigi, originally identified in a patient with recurrent venous thromboembolism, causes dysplasminogenemia with reduced plasmin activity. The Plg-A620T mutation is present in 3-4% of individuals in East Asian populations, and as many as 50,000 Japanese are estimated to be homozygous for the mutant 620T allele. In the present study, to understand the changes of thrombotic phenotypes in individuals with the mutant 620T allele, we generated knock-in mice carrying the homozygous Plg-A622T mutation (PlgT/T), an equivalent to the A620T mutation in human Plg. PlgT/T mice grew normally but showed severely reduced plasmin activity activated by urokinase, equivalent to ~8% of that in wild-type mice. In vitro fibrin clot lysis in plasma was significantly slower in PlgT/T mice than in wild-type mice. However, all experimental models of electrolytic deep vein thrombosis, tissue factor-induced pulmonary embolism, transient focal brain ischaemic stroke, or skin-wound healing showed largely similar phenotypes between PlgT/T mice and wild-type mice. Protein S-K196E mutation (Pros1E/E) is a race-specific genetic risk factor for venous thromboembolism. Coexistence in mice of PlgT/T and Pros1E/E did not affect pulmonary embolism symptoms, compared with those in Pros1E/E mice. Hence, the present study showed that the Plg-A622T mutation, which confers ~8% plasmin activity, does not increase the risk of thrombotic diseases in mice under experimental thrombotic conditions and does not modify the thrombotic phenotype observed in Pros1E/E mice. PlgT/T mice can be used to investigate the potential pathophysiological impact of the Plg-A620T mutation.


Subject(s)
Conjunctivitis/genetics , Gene Knock-In Techniques , Mutation , Phenotype , Plasminogen/deficiency , Plasminogen/genetics , Skin Diseases, Genetic/genetics , Venous Thromboembolism/genetics , Amino Acid Substitution , Animals , Brain Ischemia/blood , Brain Ischemia/genetics , Brain Ischemia/pathology , Conjunctivitis/blood , Conjunctivitis/pathology , Disease Models, Animal , Female , Fibrin/genetics , Fibrin/metabolism , Fibrinolysin/genetics , Fibrinolysin/metabolism , Gene Expression , Humans , Male , Mice , Mice, Transgenic , Plasminogen/metabolism , Protein S/genetics , Protein S/metabolism , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/pathology , Skin Diseases, Genetic/blood , Skin Diseases, Genetic/pathology , Stroke/blood , Stroke/genetics , Stroke/pathology , Venous Thromboembolism/blood , Venous Thromboembolism/pathology , Venous Thrombosis/blood , Venous Thrombosis/genetics , Venous Thrombosis/pathology , Wound Healing/physiology
3.
PLoS One ; 12(1): e0170829, 2017.
Article in English | MEDLINE | ID: mdl-28122057

ABSTRACT

Intracellular Ca2+ signaling regulates diverse functions of the nervous system. Many of these neuronal functions, including learning and memory, are regulated by neuronal calcium sensor-1 (NCS-1). However, the pathways by which NCS-1 regulates these functions remain poorly understood. Consistent with the findings of previous reports, we revealed that NCS-1 deficient (Ncs1-/-) mice exhibit impaired spatial learning and memory function in the Morris water maze test, although there was little change in their exercise activity, as determined via treadmill-analysis. Expression of brain-derived neurotrophic factor (BDNF; a key regulator of memory function) and dopamine was significantly reduced in the Ncs1-/- mouse brain, without changes in the levels of glial cell-line derived neurotrophic factor or nerve growth factor. Although there were no gross structural abnormalities in the hippocampi of Ncs1-/- mice, electron microscopy analysis revealed that the density of large dense core vesicles in CA1 presynaptic neurons, which release BDNF and dopamine, was decreased. Phosphorylation of Ca2+/calmodulin-dependent protein kinase II-α (CaMKII-α, which is known to trigger long-term potentiation and increase BDNF levels, was significantly reduced in the Ncs1-/- mouse brain. Furthermore, high voltage electric potential stimulation, which increases the levels of BDNF and promotes spatial learning, significantly increased the levels of NCS-1 concomitant with phosphorylated CaMKII-α in the hippocampus; suggesting a close relationship between NCS-1 and CaMKII-α. Our findings indicate that NCS-1 may regulate spatial learning and memory function at least in part through activation of CaMKII-α signaling, which may directly or indirectly increase BDNF production.


Subject(s)
Neuronal Calcium-Sensor Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Signal Transduction/physiology , Spatial Learning/physiology , Spatial Memory/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dopamine/metabolism , Electric Stimulation , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Mice , Mice, Knockout , Motor Activity/physiology , Neuronal Calcium-Sensor Proteins/genetics , Neuropeptides/genetics
4.
Blood ; 126(19): 2247-53, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26251307

ABSTRACT

Protein S (PS) acts as an anticoagulant cofactor for activated protein C in regulation of blood coagulation. The K196E mutation in PS is a race-specific genetic risk factor for venous thromboembolism with a prevalence of ∼2% within the Japanese population. To evaluate the thrombosis risk of the PS-K196E mutation, we generated PS-K196E knockin mice and heterozygous PS-deficient mice. We analyzed their thrombotic states, comparing with mice carrying the factor V Leiden mutation (FV-R504Q), a race-specific genetic risk for venous thrombosis in whites. PS-K196E mice grew normally but had decreased activated protein C cofactor activity in plasma. Purified recombinant murine PS-K196E showed the same decreased activated protein C cofactor activity. A deep vein thrombosis model of electrolytic inferior vena cava injury and pulmonary embolism models induced by infusion of tissue factor or polyphosphates revealed that PS-K196E mice, heterozygous PS-deficient mice, and FV-R504Q mice were much more susceptible to venous thrombosis compared with wild-type mice. Transient middle cerebral artery ischemia-reperfusion injury model studies demonstrated that both PS-K196E mice and heterozygous PS-deficient mice had cerebral infarction similar to wild-type mice, consistent with human observations. Our in vitro and in vivo results support a causal relationship between the PS-K196E mutation and venous thrombosis and indicate that PS-K196E mice can provide an in vivo evaluation system to help uncovering racial differences in thrombotic diseases.


Subject(s)
Mutation, Missense , Protein S , Venous Thromboembolism , Amino Acid Substitution , Animals , Disease Models, Animal , Factor V/genetics , Factor V/metabolism , Humans , Mice , Mice, Mutant Strains , Protein C/genetics , Protein C/metabolism , Protein S/genetics , Protein S/metabolism , Venous Thromboembolism/blood , Venous Thromboembolism/genetics
5.
Brain Res ; 1610: 110-23, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25842373

ABSTRACT

PURPOSES: An anti-allergic and analgesic drug, "an extract derived from the inflamed cutaneous tissue of rabbits inoculated with vaccinia virus (ERV)", has been used in medical practice in Japan and some other countries. We examined the effect of ERV, prior to induction of ischemia, on the development of cerebral infarction, on learning and memory, or on brain-derived neurotrophic factor (BDNF) levels in C57BL/6J mice. METHODS: Following oral administration of ERV (the same in humans: ×1) or vehicle, daily for three consecutive weeks, temporary focal ischemia was induced by the three vessel occlusion technique. In the other group of animals, after daily ERV (Low: ×1; Med: ×3, or High dose: ×9) or vehicle administration for three weeks, we performed a quantitative assessment of spatial learning or intracerebral BDNF levels. RESULTS: The volumes of infarcted lesions, brain edema and the extent of the neurological deficits were significantly reduced in the ERV-treated group. ERV treatment also enhanced spatial learning, accompanied by upregulated BDNF in the cortex. CONCLUSIONS: Daily oral intake of ERV, at a clinically relevant dose, protects the brain from ischemic stroke, and also enhances the learning function in normal mice. As millions of people are currently taking the drug safely, and have been for many years in some cases, there is a need to test the inhibitory actions of the drug on progressive dementia encountered in humans with recurrent ischemic attacks or Alzheimer's disease.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/drug effects , Cerebral Infarction/prevention & control , Neuroprotective Agents/pharmacology , Polysaccharides/pharmacology , Spatial Learning/drug effects , Acute Disease , Administration, Oral , Animals , Brain/metabolism , Brain/pathology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/prevention & control , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Chronic Disease , Cohort Studies , Disease Models, Animal , Exploratory Behavior/drug effects , Male , Memory , Mice, Inbred C57BL , Motor Activity/drug effects , Nootropic Agents/pharmacology , Random Allocation , Stroke/metabolism , Stroke/pathology , Stroke/prevention & control , Up-Regulation/drug effects
6.
Eur Neurol ; 71(1-2): 4-18, 2014.
Article in English | MEDLINE | ID: mdl-24525475

ABSTRACT

BACKGROUND/AIM: A longer period of vessel occlusion reduces the coefficient of variation of the infarct lesion size ['infarct variation coefficient' (IVC)] due to a gradual expansion of the lesion within a limited territory defined by the vascular anatomy, but it increases the mortality rate. A crucial issue in the induction of experimental focal cerebral ischemia has been to achieve a low IVC value and a low mortality rate. We attempted to improve IVC and mortality using the 3-vessel occlusion model. METHODS: We introduced a new, transtemporal fascia approach to expose the zygomatic arch, in which the fascia of the temporal muscle is cut and retracted dorsally together with the facial nerve and the vein en bloc. RESULTS/CONCLUSION: The approach avoided traumatic venous bleeding around the zygomatic arch. We established a bloodless model of focal ischemia that can produce a consistent degree of reduction in the regional cerebral blood flow within the ischemic penumbra. The model, characterized by a 15-min ischemia, an IVC of 15-21%, and low mortality after ischemia, is expected to produce reliable preclinical evidence in the assessment of neuroprotective interventions for ischemic stroke. The entire procedure is presented in the online supplementary video (www. karger.com/doi/10.1159/000356048).


Subject(s)
Disease Models, Animal , Stroke , Acute Disease , Animals , Blood Loss, Surgical/prevention & control , Brain/pathology , Brain/physiopathology , Brain/surgery , Brain Ischemia , Cerebrovascular Circulation/physiology , Chronic Disease , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Neuropsychological Tests , Neurosurgical Procedures , Survival Analysis , Video Recording
7.
Front Neurol Neurosci ; 32: 129-38, 2013.
Article in English | MEDLINE | ID: mdl-23859972

ABSTRACT

Development of a safe method to increase brain-derived neurotrophic factor (BDNF) levels in the brain is expected to enhance learning and memory, induce tolerance to cerebral infarction or tolerance to depressive state, improve glucose metabolism, and suppress appetite and body weight. We have shown that repetitive applications of high-voltage electric potential (HELP) to the body increase BDNF levels in the brain, improving learning and memory in mice. Here, we investigated the effects of HELP treatment for a chronic period on the BDNF levels in the mouse brain, and on body weight in mice and humans. Adult mice were exposed to 3.1 or 5.4 kV HELP (on the body), 5 h a day for 24 weeks, and BDNF levels in the brain and alterations in body weight were analyzed. Humans [age, 53.2 ± 15.5 years old; BMI, 27.8 ± 5.6 (mean ± SD, n = 6)] were exposed to 3.9 kV HELP (on the body) for 1 h a day, continuing for 33 months (2.8 years) under the monitor of body weight. In mice, the HELP application elevated BDNF levels in the brain at least temporarily, affecting body weight in a voltage- and time-dependent manner. In humans, the HELP treatment reduced body weight compared to the pretreated initial values without any aversive effects (p < 0.002, one-way ANOVA with the post hoc Holm-Sidak test). The results in mice indicated that 3.1 kV HELP was considered insufficient for a continuous elevation of intracerebral BDNF, and 5.4 kV HELP was considered as excessive. HELP with an appropriate voltage can be utilized to increase BDNF levels in the brain for a prolonged period. We anticipate further investigations to clarify the effect of the optimal-leveled HELP therapy on memory disturbances, neurological deficits after stroke, depression, diabetes, obesity and metabolic syndrome.


Subject(s)
Body Weight/physiology , Brain Chemistry/physiology , Brain-Derived Neurotrophic Factor/biosynthesis , Electric Stimulation/methods , Adult , Aged , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/physiology , Evoked Potentials/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
8.
Brain Res ; 1517: 104-13, 2013 Jun 23.
Article in English | MEDLINE | ID: mdl-23602966

ABSTRACT

Effective interventions that provide obvious neuroprotection are currently fairly limited. Glucagon-like peptide-1 (GLP-1), an enhancer of insulin production with a trophic effect on ß cells in the islets, has been found to be trophic for neuronal cells. Alogliptin benzoate (AGL), a selective inhibitor of dipeptidylpeptidase-4 (DPP-4) functioning as a long-acting agonist of GLP-1, is in clinical use worldwide for patients with diabetes mellitus type 2. To clarify whether administration of AGL, independent of the insulinotropic effect, protects the brain against focal ischemia, we investigated the effect of AGL on the development of cerebral infarction in non-diabetic normal mice. Male C57BL/6J mice were administered AGL (7.5, 15, or 30µg) once a day for three weeks by intragastric gavage. After the induction of temporary focal ischemia, volumes of infarcted lesions and neurological deficits were analyzed at 24h (acute phase) and seven days (chronic phase). In the acute phase, significant reductions were observed in the volumes of infarcted lesions (p=0.009), and in the severity of neurological deficits (p=0.004), in the group treated with 15µg of alogliptin benzoate, but not the 7.5 or 30µg-treated groups. This significant reduction in volumes of infarcted lesions persisted into the chronic phase. At the end of the AGL treatment; before the induction of ischemia, the levels of brain-derived neurotrophic factor (BDNF), a potent neuroprotectant in the brain, were elevated in the cortex (p=0.008), or in the whole forebrain (p=0.023). AGL could be used as a daily neuroprotectant or an enhancer of BDNF production aiming to attenuate cerebral injuries, for the growing number of people who have the risk of ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/prevention & control , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Piperidines/therapeutic use , Uracil/analogs & derivatives , Analysis of Variance , Animals , Brain Edema/etiology , Brain Edema/prevention & control , Brain Infarction/etiology , Brain Infarction/prevention & control , Brain Ischemia/complications , Brain Ischemia/pathology , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Microcirculation/physiology , Nervous System Diseases/etiology , Nervous System Diseases/prevention & control , Prosencephalon/blood supply , Prosencephalon/drug effects , Prosencephalon/metabolism , Time Factors , Uracil/therapeutic use
9.
Eur Neurol ; 68(6): 329-43, 2012.
Article in English | MEDLINE | ID: mdl-23095669

ABSTRACT

Similar to atherosclerosis, platelet-derived growth factor (PDGF)-BB, a major growth factor for vascular smooth muscle cells, is produced in arterial walls to repair arteries after subarachnoid hemorrhage (SAH). On review of a series of research articles that focus on defensive host responses to SAH, PDGF-BB is identified as a spasmogen, based on the following findings: (1) foreign substances injected into the subarachnoid space cause persistent constriction of cerebral arteries with a time course and histological features almost identical to those seen after SAH; (2) persistent constriction induced by SAH or a foreign substance is dependent on the complement system; (3) the complement system, which stimulates platelets, macrophages and endothelial cells to secrete PDGF-BB, is activated in both the cerebrospinal fluid (CSF) and plasma immediately after SAH; (4) PDGF-BB levels in the CSF are significantly elevated in patients with delayed cerebral ischemia; (5) the immunodensity of PDGF-BB in the arterial walls correlates well with the severity of cerebral vasospasm; (6) intracisternal injection of PDGF-BB induces persistent constriction of cerebral arteries in a dose-dependent manner; (7) prolonged contact with blood clots promotes the contractile response of cerebral arteries to PDGF-BB, and (8) administration of an antagonist of PDGF-BB function suppresses the development of cerebral vasospasm.


Subject(s)
Atherosclerosis/metabolism , Platelet-Derived Growth Factor/metabolism , Subarachnoid Hemorrhage/metabolism , Vasospasm, Intracranial/metabolism , Animals , Atherosclerosis/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Humans , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/cerebrospinal fluid , Platelet-Derived Growth Factor/pharmacology , Subarachnoid Hemorrhage/physiopathology , Vasospasm, Intracranial/physiopathology
10.
PLoS One ; 7(3): e34298, 2012.
Article in English | MEDLINE | ID: mdl-22479592

ABSTRACT

Accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) causes a cellular condition called ER stress. To overcome ER stress, unfolded proteins are eliminated by an ER-associated degradation (ERAD) system. To explore the physiological requirements for ERAD-related membrane proteins in mammals, we generated Derlin-1-, Derlin-3-, and Herp-deficient mice by gene targeting. Complete loss of Derlin-1 caused embryonic lethality at around E7-E8 (early somite stages). In contrast, Derlin-3- and Herp-deficient mice were born alive with the expected Mendelian frequency, and were superficially indistinguishable from wild-type mice. However, in the Derlin-3- and Herp-deficient mouse organs, the expression levels of ERAD-related proteins were affected under both normal and ER stress conditions; specific effects differed among the organs. Degradation of ERAD substrates was reduced in the Herp-deficient liver, and Herp-deficient mice exhibited impaired glucose tolerance and vulnerability to brain ischemic injury, both of which are known to be implicated in ER stress. Our findings indicate that ERAD or uncharacterized functions involving Derlin-1 are essential in early embryonic development. Derlin-3- and Herp-deficient mice may become useful model animals for investigations of the physiological contribution of ERAD under stressful or pathological conditions.


Subject(s)
Glucose/metabolism , Ischemia/metabolism , Membrane Proteins/metabolism , Animals , Brain Infarction , Crosses, Genetic , Female , Genotype , Glucose Tolerance Test , Heterozygote , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Models, Genetic , Phenotype , Protein Denaturation , Protein Folding
11.
J Biol Chem ; 286(29): 26158-65, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21636852

ABSTRACT

The N-myc downstream-regulated gene (NDRG) family consists of four related proteins, NDRG1-NDRG4, in mammals. We previously generated NDRG1-deficient mice that were unable to maintain myelin sheaths in peripheral nerves. This condition was consistent with human hereditary motor and sensory neuropathy, Charcot-Marie-Tooth disease type 4D, caused by a nonsense mutation of NDRG1. In contrast, the effects of genetic defects of the other NDRG members remain unknown. In this study, we focused on NDRG4, which is specifically expressed in the brain and heart. In situ mRNA hybridization on the brain revealed that NDRG4 was expressed in neurons of various areas. We generated NDRG4-deficient mice that were born normally with the expected Mendelian frequency. Immunochemical analysis demonstrated that the cortex of the NDRG4-deficient mice contained decreased levels of brain-derived neurotrophic factor (BDNF) and normal levels of glial cell line-derived neurotrophic factor, NGF, neurotrophin-3, and TGF-ß1. Consistent with BDNF reduction, NDRG4-deficient mice had impaired spatial learning and memory but normal motor function in the Morris water maze test. When temporary focal ischemia of the brain was induced, the sizes of the infarct lesions were larger, and the neurological deficits were more severe in NDRG4-deficient mice compared with the control mice. These findings indicate that NDRG4 contributes to the maintenance of intracerebral BDNF levels within the normal range, which is necessary for the preservation of spatial learning and the resistance to neuronal cell death caused by ischemic stress.


Subject(s)
Brain Ischemia/metabolism , Learning Disabilities/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Spatial Behavior/physiology , Amino Acid Sequence , Animals , Brain/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Circulation/genetics , Disease Susceptibility , Female , Gene Expression Regulation , Humans , Learning Disabilities/genetics , Male , Mice , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Phenotype , Protein Transport
12.
Neurosurgery ; 66(4): 728-35; discussion 735, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20305494

ABSTRACT

OBJECTIVE: After subarachnoid hemorrhage (SAH), platelet-derived growth factor-BB (PDGF-BB) is secreted in and around the cerebral arteries. To clarify the role of PDGF-BB in the development of vasospasm after SAH, we determined whether PDGF-BB alone can cause long-lasting vasoconstriction of a severity similar to that of vasospasm. In addition, the anti-vasospastic effect of trapidil, an antagonist of PDGF-BB function, was investigated. METHODS: We infused recombinant PDGF-BB (10 microg/mL saline as the vehicle) (n = 14) into the subarachnoid space of rabbits and analyzed alterations in the caliber of the basilar artery using repeated angiography. To study the role of PDGF-BB on the development of vasospasm, trapidil was administered continuously starting 1 hour after SAH, on day 0 (0.63-1.25 mg/kg /h or vehicle) for 47 hours (n = 24), or after the full development of cerebral vasospasm on day 2 (3.0 mg/kg/h or vehicle) for 0.5 hours (n = 17), and alterations in the caliber of the basilar artery were monitored. RESULTS: PDGF-BB caused long-lasting vasoconstriction, with maximum constriction of 56% (P < .001) of the control value (= 100%) on day 2, resembling vasospasm seen after SAH. Prolonged administration of intravenous trapidil, starting soon after SAH, prevented the development of vasospasm in a dose-dependent manner (P < .05, .01, or .001). Intravenous or intra-arterial administration of trapidil significantly dilated vasospasm (P < .01) on day 2, at least transiently. CONCLUSION: PDGF-BB, a growth factor synthesized in the subarachnoid space after SAH, can cause severe and long-lasting vasoconstriction. Significant prevention and resolution of vasospasm can be achieved by the PDGF-BB antagonist trapidil. We propose that excessive production of PDGF-BB, essentially aiming to repair injured arteries, causes cerebral vasospasm. Although the half-life of trapidil in serum may be shorter than that of PDGFG-BB-derived spasmogenic signaling, trapidil is a candidate drug for constructing a new therapeutic modality for preventing and resolving vasospasm.


Subject(s)
Cerebral Arteries/drug effects , Platelet-Derived Growth Factor/adverse effects , Platelet-Derived Growth Factor/metabolism , Subarachnoid Hemorrhage/physiopathology , Vasoconstriction/drug effects , Vasospasm, Intracranial/etiology , Animals , Cerebral Angiography/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Rabbits , Subarachnoid Hemorrhage/chemically induced , Time Factors , Trapidil/adverse effects , Vasodilator Agents/adverse effects
13.
Stroke ; 40(11): e606-13, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19797181

ABSTRACT

BACKGROUND AND PURPOSE: New immature neurons appear out of the germinative zone, in cortical Layers V to VI, after induced spreading depression in the adult rat brain. Because neural progenitors have been isolated in the cortex, we set out to determine whether a subgroup of mature cells in the adult cortex has the potential to divide and generate neural precursors. METHODS: We examined the expression of endogenous markers of mitotic activity, proliferating cell nuclear antigen, and vimentin as a marker for neuronal progenitor cells, if any, in the adult rat cortex after spreading depression stimulation. Immunohistochemical analysis was also performed using antibodies for proliferating cell nuclear antigen, for vimentin, and for nestin. Nestin is a marker for activity dividing neural precursors. RESULTS: At the end of spreading depression (Day 0), glial fibrillary acidic protein-positive cells in the subpial zone and cortical Layer I demonstrated increased mitotic activity, expressing vimentin and nestin. On Day 1, nestin(+) cells were found spreading in deeper cortical layers. On Day 3, vimentin(-)/nestin(+), neural precursor-like cells appeared in cortical Layers V to VI. On Day 6, new immature neurons appeared in cortical Layers V to VI. Induced spreading depression evokes cell division of astrocytes residing in the subpial zone, generating neural precursor-like cells. CONCLUSIONS: Although neural precursor-like cells found in cortical Layers V to VI might have been transferred from the germinative zone rather than the cortical subpial zone, astrocytic cells in the subpial zone may be potent neural progenitors that can help to reconstruct impaired central nervous system tissue. Special caution is required when observing or treating spreading depression waves accompanying pathological conditions in the brain.


Subject(s)
Astrocytes/cytology , Cell Division/physiology , Cerebral Cortex/cytology , Cortical Spreading Depression/physiology , Neurons/cytology , Stem Cells/cytology , Age Factors , Animals , Astrocytes/physiology , Cerebral Cortex/physiology , Male , Neurogenesis/physiology , Neurons/physiology , Pia Mater/cytology , Pia Mater/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/physiology
14.
Brain Res ; 1241: 103-9, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18801341

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a neurotrophin, is known to promote neuronal differentiation stimulating neurite outgrowth in the developing CNS, and is also known to modulate synaptic plasticity, thereby contributing to learning and memory in the mature brain. Here, we investigated the role of increased levels of intracerebral BDNF in learning and memory function. Using genetically engineered transgenic BDNF overexpressing mice (RTG-BDNF), young adult, homozygous (+/+), heterozygous (+/-), or wild-type (-/-) littermates, we analyzed escape latency to a hidden-platform and swimming velocity in the Morris Water Maze test (MWM) with modifications for the mice. The MWM comprised 4 trials per day over 5 consecutive days (sessions) without prior or subsequent training. In a separate set of animals, BDNF protein levels in the cortex, thalamostriatum and the hippocampus were measured quantitatively using ELISA. In the BDNF (+/-) mice, the BDNF levels in the cortex, the thalamostriatum and the hippocampus were significantly high, compared to the wild-type littermates; 238%, 158%, and 171%, respectively (P<0.01, one-way ANOVA and a post-hoc test in each region). The BDNF levels in the BDNF (+/+) mice were not elevated. The BDNF (+/-), but not the (+/+) mice, demonstrated significantly shorter escape latency, shorter total path length in the MWM, and more frequent arrivals at the location where the platform had been placed previously in the probe trial, compared with the wild-type littermates (P<0.05, at each time pint). Because the maximum swimming velocity was not affected in the BDNF-transgenic mice, increased BDNF levels in the brain were found to enhance spatial learning and memory function. Although it has been postulated that excessive BDNF is deteriorating for neuronal survival or neurite outgrowth, further investigations are needed to clarify the mechanism of paradoxical lack of increase in BDNF levels in the (+/+) mouse brain.


Subject(s)
Brain Chemistry/genetics , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Learning/physiology , Memory/physiology , Animals , Behavior, Animal/physiology , Brain/anatomy & histology , Cell Differentiation/genetics , Cell Survival/genetics , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/genetics , Neuropsychological Tests , Prosencephalon/anatomy & histology , Prosencephalon/metabolism , Up-Regulation/genetics
15.
Brain Res ; 1212: 79-88, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18439988

ABSTRACT

Development of a safe method to increase brain-derived neurotrophic factor (BDNF) in the brain is expected to have utility in enhancing learning and memory, in protecting the brain, and in suppressing appetite. We investigated the effects of whole-body exposure to high voltage electric potential (HELP), which generates an electric field and current density in the body, on BDNF levels in the brain, spatial learning, or resistance to cerebral infarction development after focal ischemia. Adult mice (C57BL/6J) were exposed to 3.5 kV, or 5.8 kV for 5 h a day, making indirect contact with the ground via room air, over 1, 3, 6 or 12 consecutive weeks. After treatment, BDNF levels, performances in the Morris water maze task (MWM), or development of infarct lesion after focal ischemia was analyzed. Treatment with 3.5 kV for 1, 3, 6 or 12 weeks, or with 5.8 kV for 1, 3 or 12 weeks increased BDNF levels in the cortex (P<0.05, one-way ANOVA). Every HELP treatment differentially improved escape latency in the MWM, compared with the corresponding untreated controls (P<0.05, one-way ANOVA). Treatment with 3.5 kV for 6 or 12 weeks, but not with 5.8 kV protected the brain suppressing cerebral infarction development (P<0.05). The HELP treatment with 3.5 kV for 6 or 12 weeks improves spatial learning, gently suppressing body weight gain, and protects the brain against cerebral infarction.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/physiology , Brain/radiation effects , Cerebral Infarction/prevention & control , Electromagnetic Fields/adverse effects , Learning/radiation effects , Spatial Behavior/radiation effects , Analysis of Variance , Animals , Blood Pressure/radiation effects , Cerebral Infarction/etiology , Cerebrovascular Circulation/radiation effects , Dose-Response Relationship, Radiation , Heart Rate/radiation effects , Ischemia/complications , Male , Maze Learning/radiation effects , Mice , Mice, Inbred C57BL , Swimming , Time Factors
17.
Stroke ; 36(7): 1544-50, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15933264

ABSTRACT

BACKGROUND AND PURPOSE: Status epilepticus and cerebral ischemia stimulate persistent neurogenesis in the adult brain, but both conditions cause neuronal damage. We determined whether spreading depression, a common epiphenomenon of these conditions, stimulates persistent neurogenesis. METHODS: We analyzed the effect of KCl-induced spreading depression on persistent neurogenesis and the spatio-temporal distribution of cells exhibiting immunohistochemical markers for divided and early committed neurons (new neurons) in the adult rat brain. RESULTS: After induction of spreading depression for 48 hours, the density of mitotic cells, divided cells, and new neurons in the subventricular zone increased at days 1 to 3, days 3 to 6, and day 6, respectively (P<0.05). The divided cell density in the rostral migratory stream and the stream size increased at day 12 (P<0.001). Vehicle (saline) infusion or induction of spreading depression for 4 hours only did not increase the divided cell density, but the latter increased new neuron density in the subventricular zone (P<0.001). Double-labeled new neuron-like cells also appeared in the caudate putamen or cortex in ectopic fashion at day 3, with dramatic increases at days 6 and 12. Administration of the NMDA receptor antagonist, MK-801, which inhibits the propagation of spreading depression, abolished the increase in new neurons in the subventricular zone and the appearance of ectopic new neuron-like cells after 48-hour KCl infusion. There was no neuronal damage, as evidenced by mature neuron density, neurite density, and apoptotic cell appearance after spreading depression for 48 hours. CONCLUSIONS: Spreading depression has the potential to stimulate persistent neurogenesis or to produce ectopic new neuron-like cells.


Subject(s)
Biomarkers/chemistry , Cerebral Cortex/metabolism , Cortical Spreading Depression , Neurons/pathology , Putamen/metabolism , Animals , Antimetabolites/pharmacology , Apoptosis , Brain/metabolism , Brain Ischemia/pathology , Bromodeoxyuridine/pharmacology , Cell Differentiation , Cell Division , Central Nervous System/pathology , Dizocilpine Maleate/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Membrane Potentials , Microscopy, Confocal , Neurons/metabolism , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Status Epilepticus/pathology , Stem Cells/metabolism , Time Factors
18.
Brain Res ; 1019(1-2): 178-88, 2004 Sep 03.
Article in English | MEDLINE | ID: mdl-15306252

ABSTRACT

Preconditioning the rat brain with spreading depression for 48 h induces potent ischemic tolerance (infarct tolerance) after an interval of 12-15 days, consequently reducing the infarcted lesion size in the acute phase following focal cerebral ischemia. However, persistence of the morphological and functional neuroprotection has not yet been proven. We tested whether tolerance-derived neuroprotection against focal cerebral ischemia persists or merely delays the progress of cerebral infarction. Prolonged spreading depression was induced in mice by placing a depolarized focus with intracerebral microinfusion of KCl for 24 h; after intervals of 3, 6, 9 or 12 days, temporary focal ischemia was imposed. In the analysis of the infarcted lesion volume 24 h after ischemia, groups with 6 or 9 day interval demonstrated significantly smaller lesion volume compared to time-matched vehicle control group (P=0.002). Significant reduction in cerebral infarction was also observed at the chronic phase, namely 14 days after ischemia (33% reduction) (P=0.021) accompanied with less severe neurological deficits (38% reduction) (P=0.020). Using this technique, we also investigated if the mice with targeted disruption of a single BDNF allele (heterozygous BDNF-deficient mice) can gain the same potency of tolerance as the wild mice. In the result on infarcted lesion volumes following temporary focal ischemia, potent tolerance developed in the wild type (35% reduction) (P=0.007) but not in the heterozygous BDNF-deficient mice (<19% reduction) (P=0.155), indicating that BDNF expression level following spreading depression is contributing to infarct tolerance development.


Subject(s)
Brain Infarction/genetics , Brain Infarction/pathology , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Cortical Spreading Depression/genetics , Animals , Brain Infarction/prevention & control , Brain-Derived Neurotrophic Factor/biosynthesis , Female , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors
19.
Exp Neurol ; 182(2): 261-74, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12895438

ABSTRACT

The temporary three-vessel occlusion (3VO) technique with a surgical approach for middle cerebral artery (MCA) produces consistent cerebral infarction in the neocortex in normotensive rats. The intraluminal thread-occlusion technique with an endovascular approach targeting the MCA occlusion (MCAO) is more widely used since it does not require complicated intracranial procedures. The aim of this study was to review the methods/models for MCAO stroke in normotensive rats and to evaluate a 3VO stroke model that provides consistent degrees and variance of cortical stroke injury for additional discussion. First, we analyzed a model with modified temporary 3VO technique requiring less complicated procedures than the temporary 3VO model, i.e., temporary occlusion of the bilateral common carotid arteries (CCAs) superimposed on a permanent occlusion of the MCA, in Sprague-Dawley rats or C57BL/6J mice. In the microvascular tissue (cerebral) perfusion study, significant reductions in regional cerebral perfusion during the 3VO accompanied a rapid return to baseline after release of the CCAs, showing that the technique induces temporary focal ischemia. The average sizes and variances of the neocortical infarction in this model, together with those in the other normotensive rat models caused by the 3VO technique in the literature, indicated a standard size and variance of infarcted lesion in the control groups relative to the specific ischemic period. However, stroke injuries in the neocortex induced by the thread occlusion technique showed greater variability with less consistent lesion sizes. Inclusion/exclusion criteria to avoid inappropriate cases with too mild (no/faint infarction) or too great (huge/fatal infarction) severity in the ischemic injury may differ between laboratories in the thread occlusion model.


Subject(s)
Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Neocortex/pathology , Stroke/pathology , Vascular Surgical Procedures/methods , Animals , Blood Pressure , Infarction, Middle Cerebral Artery/complications , Ischemic Attack, Transient/pathology , Mice , Mice, Inbred C57BL , Neocortex/blood supply , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Stroke/complications , Vascular Surgical Procedures/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...