Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
2.
BMC Neurol ; 24(1): 32, 2024 Jan 18.
Article En | MEDLINE | ID: mdl-38238659

BACKGROUND: Although randomized controlled trials (RCTs) have shown that calcitonin gene-related peptide (CGRP)-targeted monoclonal antibodies (CGRP mAbs) are an efficacious and safe therapeutic modality for migraine prevention, their clinical benefits have not been well validated in Japanese patients in the real-world setting. The present study aimed to evaluate the real-world efficacy and safety of galcanezumab, fremanezumab, and erenumab in Japanese patients with migraine. METHODS: This observational retrospective cohort study was conducted at two headache centers in Japan. Patients with migraine who had experienced treatment failure with at least one traditional oral migraine preventive agent were treated with a CGRP mAb de novo. The primary efficacy endpoints were the changes from baseline in monthly migraine days (MMDs) and Headache Impact Test-6 (HIT-6) score after 3 dosing intervals (V3). We explored whether demographic and clinical characteristics predicted therapeutic outcomes at V3. RESULTS: Sixty-eight patients who completed three doses of a CGRP mAb (85.3% female [58/68], mean age: 46.2 ± 13.1 years) were included in the analysis. There were 19 patients with chronic migraine. The baseline MMDs were 13.4 ± 6.0. After 3 doses, the MMDs significantly decreased to 7.4 ± 5.5 (p < 0.0001), and the 50% response rate was 50.0%. HIT-6 score was significantly reduced from 66.7 ± 5.4 to 56.2 ± 8.7 after 3 doses (P = 0.0001). There was a positive correlation between the changes in MMDs and HIT-6 score from baseline after 2 doses (p = 0.0189). Those who achieved a ≥ 50% therapeutic response after the first and second doses were significantly more likely to do so at V3 (crude odds ratio: 3.474 [95% CI: 1.037 to 10.4], p = 0.0467). The most frequent adverse event was constipation (7.4%). None of the adverse events were serious, and there was no need for treatment discontinuation. CONCLUSIONS: This real-world study demonstrated that CGRP mAbs conferred Japanese patients with efficacious and safe migraine prevention, and an initial positive therapeutic response was predictive of subsequent favorable outcomes. Concomitant measurement of MMDs and HIT-6 score was useful in evaluating the efficacy of CGRP mAbs in migraine prevention.


Calcitonin Gene-Related Peptide , Migraine Disorders , Adult , Female , Humans , Male , Middle Aged , Cohort Studies , Headache/drug therapy , Japan/epidemiology , Migraine Disorders/drug therapy , Migraine Disorders/prevention & control
3.
Arch Gerontol Geriatr ; 96: 104439, 2021.
Article En | MEDLINE | ID: mdl-34090127

BACKGROUND: Post-stroke dysphagia is a common and expensive complication of acute stroke. The relationship between dysphagia and skeletal muscle loss (sarcopenia) has been recently highlighted. This study aimed to determine the relationship between temporal muscle thickness (TMT) measured by head magnetic resonance imaging (MRI) and dysphagia in patients with acute stroke. METHODS: Seventy participants (43 men and 27 women; mean age, 75.6 ± 12.7 years) were included in this study. TMT was measured by T2-magnetic resonance images within seven days of hospitalization. The severity of dysphagia was assessed using the Functional Oral Intake Scale (FOIS). Participants were classified into three categories according to the severity of dysphagia (severe: FOIS score, 1-3; mild: FOIS score, 4-6; normal: FOIS score, 7). Linear regression analysis was used to determine the independent explanators of dysphagia severity. RESULTS: Twenty participants (28.6%) had severe dysphagia, 31 participants (44.3%) had mild dysphagia, and 19 participants (27.1%) had normal swallowing function at discharge. The results of the linear regression analysis showed that TMT was a significant explanator of dysphagia severity following stroke, along with age and National Institute of Health Stroke Scale (NIHSS) score (P < 0.05, effect size: f2 = 0.72). CONCLUSIONS: TMT was an independent risk factor for dysphagia in patients with acute stroke. Skeletal muscle loss may be secondarily involved in dysphagia with acute stroke, and measurement of TMT with head MRI is a useful method to assess skeletal muscle loss.


Deglutition Disorders , Sarcopenia , Stroke , Aged , Deglutition , Deglutition Disorders/diagnostic imaging , Deglutition Disorders/etiology , Female , Humans , Male , Stroke/complications , Stroke/diagnostic imaging , Temporal Muscle
4.
J Neurosci ; 40(42): 8160-8173, 2020 10 14.
Article En | MEDLINE | ID: mdl-32928888

The amyloid-ß (Aß) peptide, a key pathogenic factor in Alzheimer's disease, attenuates the increase in cerebral blood flow (CBF) evoked by neural activity (functional hyperemia), a vital homeostatic response in which NMDA receptors (NMDARs) play a role through nitric oxide, and the CBF increase produced by endothelial factors. Tissue plasminogen activator (tPA), which is reduced in Alzheimer's disease and in mouse models of Aß accumulation, is required for the full expression of the NMDAR-dependent component of functional hyperemia. Therefore, we investigated whether tPA is involved in the neurovascular dysfunction of Aß. tPA activity was reduced, and the tPA inhibitor plasminogen inhibitor-1 (PAI-1) was increased in male mice expressing the Swedish mutation of the amyloid precursor protein (tg2576). Counteracting the tPA reduction with exogenous tPA or with pharmacological inhibition or genetic deletion of PAI-1 completely reversed the attenuation of the CBF increase evoked by whisker stimulation but did not ameliorate the response to the endothelium-dependent vasodilator acetylcholine. The tPA deficit attenuated functional hyperemia by suppressing NMDAR-dependent nitric oxide production during neural activity. Pharmacological inhibition of PAI-1 increased tPA activity, prevented neurovascular uncoupling, and ameliorated cognition in 11- to 12-month-old tg2576 mice, effects associated with a reduction of cerebral amyloid angiopathy but not amyloid plaques. The data unveil a selective role of the tPA in the suppression of functional hyperemia induced by Aß and in the mechanisms of cerebral amyloid angiopathy, and support the possibility that modulation of the PAI-1-tPA pathway may be beneficial in diseases associated with amyloid accumulation.SIGNIFICANCE STATEMENT Amyloid-ß (Aß) peptides have profound neurovascular effects that may contribute to cognitive impairment in Alzheimer's disease. We found that Aß attenuates the increases in blood flow evoked by neural activation through a reduction in tissue plasminogen activator (tPA) caused by upregulation of its endogenous inhibitor plasminogen inhibitor-1 (PAI-1). tPA deficiency prevents NMDA receptors from triggering nitric oxide production, thereby attenuating the flow increase evoked by neural activity. PAI-1 inhibition restores tPA activity, rescues neurovascular coupling, reduces amyloid deposition around blood vessels, and improves cognition in a mouse model of Aß accumulation. The findings demonstrate a previously unappreciated role of tPA in Aß-related neurovascular dysfunction and in vascular amyloid deposition. Restoration of tPA activity could be of therapeutic value in diseases associated with amyloid accumulation.


Amyloid beta-Protein Precursor/genetics , Blood Vessels/drug effects , Blood Vessels/physiopathology , Cerebral Amyloid Angiopathy/physiopathology , Cerebrovascular Disorders/physiopathology , Neurons/drug effects , Tissue Plasminogen Activator/deficiency , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebral Amyloid Angiopathy/genetics , Cerebrovascular Circulation , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/prevention & control , Cognition , Humans , Hyperemia/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide/biosynthesis , Physical Stimulation , Receptors, N-Methyl-D-Aspartate/metabolism , Serpin E2/genetics , Tissue Plasminogen Activator/genetics , Vibrissae/innervation
5.
Epilepsy Res ; 166: 106428, 2020 10.
Article En | MEDLINE | ID: mdl-32702537

It has been speculated that patients with epilepsy in elderly is increasing. The effect of enzyme-inducing antiepileptic drugs (EIAED) on lipid metabolism is well known. However, the study in elderly subjects has been rarely conducted. Furthermore, it has not been directly assessed whether EIAED use causes the change of lipid levels from normal range to abnormal range. Adult patients with epilepsy (235 on EIAED, 213 on non-EIAED) were divided by age (20-64, 65-74 and 75 over years old). Longitudinal change (before starting AED, in 3 and 6 months after starting AED) of serum total cholesterol (TC), triglyceride (TRIG), HDL cholesterol (HDL-C), LDL cholesterol (LDL-C) and non-HDL cholesterol (non-HDL-C) were retrospectively analyzed using mixed models. The risk of change of lipid levels from normal range to abnormal range was also assessed by logistic regression analysis. Comparison between patients with EIAED use (EIAED group) and patients with non-EIAED use (non-EIAED group) revealed significant differences. non-EIAED group was set as reference. Differences in least square mean (LSM), 95 % confidence interval (CI) and P value in analysis of LDL-C and non-HDL-C levels (1 × 10-2) were as follows: 0.23 (0.10 ∼ 0.36), P < 0.001, 0.22 (0.09 ∼ 0.33), P = 0.001 in patients with age 65-74. In patients of age over 75, those were 0.08 (0.01 ∼ 0.15), P = 0.038, 0.17 (0.07 ∼ 0.26), P < 0.001, respectively. In patients with age 65-74, odds ratios (ORs) (95 % CI) in change of TC, LDL and non-HDL-C level from normal range to abnormal range were 32.28 (1.83-567.05), 48.43 (2.69-869.05), 12.04 (1.29-111.88), respectively. In patients with age 75 over, odds ratios (ORs) (95 % CI) in change of TC, LDL and non-HDL-C level from normal range to abnormal range were 46.02 (2.77-762.89), 5.53 (1.27-23.94), 2.38 (1.01-10.76), respectively. EIAED use had significant impact on changes in lipid levels in elderly patients. Furthermore, we could assess the impact of switching AED from EIAED to levetiracetam (LEV) in 36 patients including elderly. Switching AED from EIAED to LEV had significant impact in reducing TC, TRIG, LDL-C and non-HDL-C in both younger adult and elderly patients.


Anticonvulsants/administration & dosage , Cytochrome P-450 Enzyme Inducers/administration & dosage , Epilepsy/blood , Epilepsy/drug therapy , Lipid Metabolism/drug effects , Age Factors , Aged , Aged, 80 and over , Anticonvulsants/adverse effects , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cytochrome P-450 Enzyme Inducers/adverse effects , Female , Humans , Lipid Metabolism/physiology , Longitudinal Studies , Male , Middle Aged , Retrospective Studies , Triglycerides/blood
6.
Nat Commun ; 9(1): 3816, 2018 09 19.
Article En | MEDLINE | ID: mdl-30232327

The ApoE4 allele is associated with increased risk of small vessel disease, which is a cause of vascular cognitive impairment. Here, we report that mice with targeted replacement (TR) of the ApoE gene with human ApoE4 have reduced neocortical cerebral blood flow compared to ApoE3-TR mice, an effect due to reduced vascular density rather than slowing of microvascular red blood cell flow. Furthermore, homeostatic mechanisms matching local delivery of blood flow to brain activity are impaired in ApoE4-TR mice. In a model of cerebral hypoperfusion, these cerebrovascular alterations exacerbate damage to the white matter of the corpus callosum and worsen cognitive dysfunction. Using 3-photon microscopy we found that the increased white matter damage is linked to an enhanced reduction of microvascular flow resulting in local hypoxia. Such alterations may be responsible for the increased susceptibility to hypoxic-ischemic lesions in the subcortical white matter of individuals carrying the ApoE4 allele.


Apolipoproteins E/metabolism , Cognition/physiology , White Matter/blood supply , Animals , Carotid Stenosis , Cerebrovascular Circulation , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Gene Targeting , Humans , Metalloporphyrins , Mice, Inbred C57BL , Mice, Transgenic , Rest , White Matter/pathology , White Matter/physiopathology
7.
Nat Neurosci ; 21(2): 240-249, 2018 02.
Article En | MEDLINE | ID: mdl-29335605

A diet rich in salt is linked to an increased risk of cerebrovascular diseases and dementia, but it remains unclear how dietary salt harms the brain. We report that, in mice, excess dietary salt suppresses resting cerebral blood flow and endothelial function, leading to cognitive impairment. The effect depends on expansion of TH17 cells in the small intestine, resulting in a marked increase in plasma interleukin-17 (IL-17). Circulating IL-17, in turn, promotes endothelial dysfunction and cognitive impairment by the Rho kinase-dependent inhibitory phosphorylation of endothelial nitric oxide synthase and reduced nitric oxide production in cerebral endothelial cells. The findings reveal a new gut-brain axis linking dietary habits to cognitive impairment through a gut-initiated adaptive immune response compromising brain function via circulating IL-17. Thus, the TH17 cell-IL-17 pathway is a putative target to counter the deleterious brain effects induced by dietary salt and other diseases associated with TH17 polarization.


Cerebrovascular Disorders/chemically induced , Cognition Disorders/chemically induced , Intestine, Small/pathology , Sodium Chloride, Dietary/toxicity , Th17 Cells/drug effects , Acetylcholine/pharmacology , Amides/pharmacology , Animals , Antihypertensive Agents/pharmacology , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cerebrovascular Circulation/drug effects , Cerebrovascular Disorders/drug therapy , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interleukin-17/administration & dosage , Interleukin-17/blood , Interleukin-17/genetics , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurovascular Coupling/drug effects , Phosphorylation/drug effects , Pyridines/pharmacology
8.
Circ Res ; 121(3): 258-269, 2017 Jul 21.
Article En | MEDLINE | ID: mdl-28515043

RATIONALE: Increasing evidence indicates that alterations of the cerebral microcirculation may play a role in Alzheimer disease, the leading cause of late-life dementia. The amyloid-ß peptide (Aß), a key pathogenic factor in Alzheimer disease, induces profound alterations in neurovascular regulation through the innate immunity receptor CD36 (cluster of differentiation 36), which, in turn, activates a Nox2-containing NADPH oxidase, leading to cerebrovascular oxidative stress. Brain perivascular macrophages (PVM) located in the perivascular space, a major site of brain Aß collection and clearance, are juxtaposed to the wall of intracerebral resistance vessels and are a powerful source of reactive oxygen species. OBJECTIVE: We tested the hypothesis that PVM are the main source of reactive oxygen species responsible for the cerebrovascular actions of Aß and that CD36 and Nox2 in PVM are the molecular substrates of the effect. METHODS AND RESULTS: Selective depletion of PVM using intracerebroventricular injection of clodronate abrogates the reactive oxygen species production and cerebrovascular dysfunction induced by Aß applied directly to the cerebral cortex, administered intravascularly, or overproduced in the brain of transgenic mice expressing mutated forms of the amyloid precursor protein (Tg2576 mice). In addition, using bone marrow chimeras, we demonstrate that PVM are the cells expressing CD36 and Nox2 responsible for the dysfunction. Thus, deletion of CD36 or Nox2 from PVM abrogates the deleterious vascular effects of Aß, whereas wild-type PVM reconstitute the vascular dysfunction in CD36-null mice. CONCLUSIONS: The data identify PVM as a previously unrecognized effector of the damaging neurovascular actions of Aß and unveil a new mechanism by which brain-resident innate immune cells and their receptors may contribute to the pathobiology of Alzheimer disease.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cerebrovascular Circulation/physiology , Macrophages/metabolism , Somatosensory Cortex/blood supply , Somatosensory Cortex/metabolism , Alzheimer Disease/pathology , Animals , Brain/blood supply , Brain/metabolism , Brain/pathology , Macrophages/pathology , Male , Mice , Mice, Transgenic , Random Allocation , Reactive Oxygen Species/metabolism
9.
PLoS One ; 11(9): e0163329, 2016.
Article En | MEDLINE | ID: mdl-27657726

Hypercapnia induces potent vasodilation in the cerebral circulation. Although it has long been known that prostanoids participate in the cerebrovascular effects of hypercapnia, the role of prostaglandin E2 (PGE2) and PGE2 receptors have not been fully investigated. In this study, we sought to determine whether cyclooxygenase-1 (COX-1)-derived PGE2 and EP1 receptors are involved in the cerebrovascular response induced by hypercapnia. Cerebral blood flow (CBF) was recorded by laser-Doppler flowmetry in the somatosenasory cortex of anesthetized male EP1-/- mice and wild type (WT) littermates. In WT mice, neocortical application of the EP1 receptor antagonist SC-51089 attenuated the increase in CBF elicited by systemic hypercapnia (pCO2 = 50-60 mmHg). SC-51089 also attenuated the increase in CBF produced by neocortical treatment of arachidonic acid or PGE2. These CBF responses were also attenuated in EP1-/- mice. In WT mice, the COX-1 inhibitor SC-560, but not the COX-2 inhibitor NS-398, attenuated the hypercapnic CBF increase. Neocortical application of exogenous PGE2 restored the attenuation in resting CBF and the hypercapnic response induced by SC-560. In contrast, exogenous PGE2 failed to rescue the attenuation both in WT mice induced by SC-51089 and EP1-/- mice, attesting to the obligatory role of EP1 receptors in the response. These findings indicate that the hypercapnic vasodilatation depends on COX-1-derived PGE2 acting on EP1 receptors and highlight the critical role that COX-1-derived PGE2 and EP1 receptors play in the hypercapnic regulation of the cerebral circulation.

10.
Cell Mol Neurobiol ; 36(2): 155-65, 2016 Mar.
Article En | MEDLINE | ID: mdl-26328781

Alzheimer's disease (AD) and cerebrovascular diseases share common vascular risk factors that have disastrous effects on cerebrovascular regulation. Endothelial cells, lining inner walls of cerebral blood vessels, form a dynamic interface between the blood and the brain and are critical for the maintenance of neurovascular homeostasis. Accordingly, injury in endothelial cells is regarded as one of the earliest symptoms of impaired vasoregulatory mechanisms. Extracellular buildup of amyloid-ß (Aß) is a central pathogenic factor in AD. Aß exerts potent detrimental effects on cerebral blood vessels and impairs endothelial structure and function. Recent evidence implicates vascular oxidative stress and activation of the non-selective cationic channel transient receptor potential melastatin (TRPM)-2 on endothelial cells in the mechanisms of Aß-induced neurovascular dysfunction. Thus, Aß triggers opening of TRPM2 channels in endothelial cells leading to intracellular Ca(2+) overload and vasomotor dysfunction. The cerebrovascular dysfunction may contribute to AD pathogenesis by reducing the cerebral blood supply, leading to increased susceptibility to vascular insufficiency, and by promoting Aß accumulation. The recent realization that vascular factors contribute to AD pathobiology suggests new targets for the prevention and treatment of this devastating disease.


Amyloid beta-Peptides/toxicity , Brain/blood supply , Brain/physiopathology , Endothelium, Vascular/physiopathology , Alzheimer Disease/pathology , Animals , Brain/drug effects , Brain/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Humans , Oxidative Stress/drug effects
11.
Stroke ; 45(6): 1815-21, 2014 Jun.
Article En | MEDLINE | ID: mdl-24781082

BACKGROUND AND PURPOSE: Accumulation of amyloid-ß in cerebral blood vessels occurs in familial and sporadic forms of cerebral amyloid angiopathy and is a prominent feature of Alzheimer disease. However, the functional correlates of the vascular pathology induced by cerebral amyloid angiopathy and the mechanisms involved have not been fully established. METHODS: We used male transgenic mice expressing the Swedish, Iowa, and Dutch mutations of the amyloid precursor protein (Tg-SwDI) to examine the effect of cerebral amyloid angiopathy on cerebrovascular structure and function. Somatosensory cortex cerebral blood flow was monitored by laser-Doppler flowmetry in anesthetized Tg-SwDI mice and wild-type littermates equipped with a cranial window. RESULTS: Tg-SwDI mice exhibited reductions in cerebral blood flow responses to whisker stimulation, endothelium-dependent vasodilators, or hypercapnia at 3 months when compared with wild-type mice, whereas the response to adenosine was not attenuated. However, at 18 and 24 months, all cerebrovascular responses were markedly reduced. At this time, there was evidence of cerebrovascular amyloid deposition, smooth muscle fragmentation, and pericyte loss. Neocortical superfusion with the free radical scavenger manganic(I-II)meso-tetrakis(4-benzoic acid) porphyrin rescued endothelium-dependent responses and functional hyperemia completely at 3 months but only partially at 18 months. CONCLUSIONS: Tg-SwDI mice exhibit a profound age-dependent cerebrovascular dysfunction, involving multiple regulatory mechanisms. Early in the disease process, oxidative stress is responsible for most of the vascular dysfunction, but with advancing disease structural alterations of the vasomotor apparatus also play a role. Early therapeutic interventions are likely to have the best chance to counteract the deleterious vascular effects of cerebral amyloid angiopathy.


Amyloid beta-Peptides/biosynthesis , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/physiopathology , Cerebrovascular Circulation , Somatosensory Cortex , Amyloid beta-Peptides/genetics , Animals , Blood Flow Velocity , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/pathology , Disease Models, Animal , Male , Mice , Mice, Transgenic , Somatosensory Cortex/blood supply , Somatosensory Cortex/metabolism , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Time Factors
12.
Stroke ; 45(4): 1131-8, 2014 Apr.
Article En | MEDLINE | ID: mdl-24619393

BACKGROUND AND PURPOSE: Prohibitin is a multi-functional protein involved in numerous cellular activities. Prohibitin overexpression protects neurons from injury in vitro, but it is unclear whether prohibitin can protect selectively vulnerable hippocampal CA1 neurons in a clinically relevant injury model in vivo and, if so, whether the salvaged neurons remain functional. METHODS: A mouse model of transient forebrain ischemia that mimics the brain damage produced by cardiac arrest in humans was used to test whether prohibitin expression protects CA1 neurons from injury. Prohibitin-expressing viral vector was microinjected in mouse hippocampus to upregulate prohibitin. RESULTS: Prohibitin overexpression protected CA1 neurons from transient forebrain ischemia. The protection was associated with dampened postischemic reactive oxygen species generation, reduced mitochondrial cytochrome c release, and decreased caspase-3 activation. Importantly, the improvement in CA1 neuronal viability translated into an improvement in hippocampal function: prohibitin expression ameliorated the spatial memory deficit induced by ischemia, assessed by the Y-maze test, and restored postischemic synaptic plasticity assessed by long-term potentiation, indicating that the neurons spared form ischemic damage were functionally competent. CONCLUSIONS: These data demonstrate that prohibitin overexpression protects highly vulnerable CA1 neurons from ischemic injury in vivo and suggest that the effect is mediated by reduction of postischemic reactive oxygen species generation and preservation of mitochondrial outer membrane integrity that prevents activation of apoptosis. Measures to enhance prohibitin expression could have translational value in ischemic brain injury and, possibly, other forms of brain injury associated with mitochondrial dysfunction.


CA1 Region, Hippocampal/pathology , Genetic Therapy/methods , Ischemic Attack, Transient/pathology , Repressor Proteins/genetics , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiopathology , Cell Count , Dependovirus/genetics , Disease Models, Animal , Gene Transfer Techniques , Heart Arrest/pathology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/physiopathology , Maze Learning/physiology , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mitochondria/physiology , Neurons/metabolism , Neurons/pathology , Prohibitins , Reactive Oxygen Species
13.
Neurourol Urodyn ; 32(5): 428-34, 2013 Jun.
Article En | MEDLINE | ID: mdl-23129306

AIMS: Overactive bladder (OAB) affects the daily life of many stroke victims. In contrast to urinary incontinence, little is known about the prevalence and risk factors for OAB among stroke patients. Therefore, we conducted a questionnaire survey and analyzed the results together with the clinical data and MRI findings. METHODS: A total of 500 volunteer patients with chronic-phase stroke were enrolled. The overactive bladder symptom score (OABSS), Short Form 8 (SF-8) health survey questionnaire, and some key international questionnaires about urinary dysfunction were assessed. RESULTS: We diagnosed 141 patients (28%) with OAB, among whom 103 (73%) had never been treated for their symptoms. Patients with OAB showed lower scores in both the physical and mental components of the SF-8, which suggested the burden of OAB on the quality of life of stroke patients. Advanced age and male gender were closely related to high OABSS. The modified Rankin Scale (mRS) was positively correlated with OABSS. Patients with cerebral infarction and those with intracerebral hemorrhage showed a similarly high OABSS. The severity of deep white-matter hyperintensity on MRI, classified by the 4-grade Fazekas scoring system, was significantly associated with high OABSS irrespective of presence of accompanying infarcts. Patients with cerebral infarcts in the region of anterior circulation showed a higher OABSS than those with cerebral infarcts in the posterior circulation. CONCLUSIONS: Based on the present risk analysis, patient care should be preferentially focused on the detection and treatment of OAB to improve the quality of life of stroke patients.


Quality of Life , Stroke/psychology , Urinary Bladder, Overactive/psychology , Age Factors , Aged , Aged, 80 and over , Cost of Illness , Female , Humans , Japan/epidemiology , Logistic Models , Magnetic Resonance Imaging , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Prevalence , Prognosis , Risk Factors , Severity of Illness Index , Sex Factors , Stroke/diagnosis , Stroke/epidemiology , Stroke/physiopathology , Surveys and Questionnaires , Urinary Bladder, Overactive/diagnosis , Urinary Bladder, Overactive/epidemiology , Urinary Bladder, Overactive/physiopathology
14.
Stroke ; 44(1): 198-204, 2013 Jan.
Article En | MEDLINE | ID: mdl-23204056

BACKGROUND AND PURPOSE: Amyloid-ß (Aß), a peptide that accumulates in the brain and circulates in the blood of patients with Alzheimer disease, alters the regulation of cerebral blood flow and may contribute to the brain dysfunction underlying the dementia. However, the contributions of brain and circulating Aß1-40 to the vascular dysfunction have not been elucidated. METHODS: We used transgenic mice overexpressing mutated forms of the amyloid precursor protein in which Aß1-40 is elevated in blood and brain (Tg-2576) or only in brain (Tg-SwDI). Mice were equipped with a cranial window, and the increase in cerebral blood flow induced by neural activity (whisker stimulation), or by topical application of endothelium-dependent vasodilators, was assessed by laser-Doppler flowmetry. RESULTS: The cerebrovascular dysfunction was observed also in Tg-SwDI mice, but despite ≈40% higher levels of brain Aß1-40, the effect was less marked than in Tg-2576 mice. Intravascular administration of Aß1-40 elevated plasma Aß1-40 and enhanced the dysfunction in Tg-SwDI mice, but not in Tg-2576 mice. CONCLUSIONS: The results provide evidence that Aß1-40 acts on distinct luminal and abluminal vascular targets, the deleterious cerebrovascular effects of which are additive. Furthermore, the findings highlight the importance of circulating Aß1-40 in the cerebrovascular dysfunction and may provide insight into the cerebrovascular alterations in conditions in which elevations in plasma Aß1-40 occur.


Amyloid beta-Peptides/biosynthesis , Brain/blood supply , Brain/physiopathology , Cerebrovascular Circulation/physiology , Peptide Fragments/biosynthesis , Vasomotor System/metabolism , Vasomotor System/physiopathology , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/blood
15.
Rinsho Shinkeigaku ; 52(11): 1365-8, 2012.
Article Ja | MEDLINE | ID: mdl-23196620

Among many conditions causing small vessel diseases, lipohyalinosis is the leading pathology next to microatheroma. Lipohyalinosis affects penetrating arteries distally than microatheroma. Proliferation of smooth muscle cells may occlude the lumen, reducing the blood flow and inducing lacunar infarction. In contrast, fibrinoid necrosis of smooth muscle cells in the media may weaken the vascular constriction, increasing the perfusion pressure in the capillary and damaging the blood brain barrier which can induce white matter lesion. Neurovascular unit (NVU) is a concept that neurons, astrocytes, and vessels function as a unit to support neuronal activity. NVU is involved in the maintenance of synapse, transmitter, energy metabolism, blood-brain barrier, and blood flow. Change in neuronal activity is transmitted to capillaries through NVU, where the information is collected along vessels proximally and regulates blood flow (proximal integration model). CARASIL and CADASIL both affect vascular smooth muscle cells, resulting in vascular dilatation, damaging NVU, and inducing white matter lesion. Occlusion of the affected vessels, causing cerebral ischemia, under these diseases is relatively infrequent. Similarity in pathophysiology between hypertensive arteriolar disease and the familial angiopathy may indicate that injury to NVU may indicate the common pathophysiology of white matter lesions.


Alopecia/pathology , CADASIL/pathology , Cerebral Infarction/pathology , Cerebrovascular Disorders/pathology , Leukoencephalopathies/pathology , Spinal Diseases/pathology , Alopecia/physiopathology , CADASIL/physiopathology , Cerebral Infarction/physiopathology , Humans , Leukoencephalopathies/physiopathology , Microcirculation/physiology , Muscle, Smooth, Vascular/pathology , Spinal Diseases/physiopathology
16.
Neurobiol Dis ; 48(3): 367-78, 2012 Dec.
Article En | MEDLINE | ID: mdl-22820141

Botulinum neurotoxin type-A (BoNT-A) is clinically used for patients with pain disorders and dystonia. The precise mechanism whereby BoNT-A controls pain remains elusive. Here, we studied how BoNT-A affects the expression of the transient receptor potential vanilloid subfamily member 1 (TRPV1), a cation channel critically implicated in nociception, in the trigeminal system. Histological studies revealed that subcutaneous BoNT-A injection (0.25, 0.5, or 5 ng/kg) into the face targeted the ophthalmic division of trigeminal ganglion (TG) neurons and decreased TRPV1-immunoreactive neurons in the TG and TRPV1-immunoreactive fibers in rat trigeminal terminals. Of note, TG neurons that received projections from the dura mater, a principal site of headache generation, had reduced TRPV1 expression. BoNT-A-induced cleavage of SNAP25 (synaptosomal-associated protein of 25-kDa) in the TG became obvious 2 days after BoNT-A administration and persisted for at least 14 days. Quantitative real-time RT-PCR (reverse transcription-polymerase chain reaction) data indicated that the TRPV1-decreasing effects of BoNT-A were not mediated by transcriptional downregulation. By employing a surface protein biotin-labeling assay, we demonstrated that BoNT-A inhibited TRPV1 trafficking to the plasma membrane in primary TG neurons. Moreover, Y200F-mutated TRPV1, which is incapable of trafficking to the plasma membrane, was expressed in PC12 cells by transfection, and pharmacological studies revealed that TRPV1 in the cytoplasm was more predisposed to proteasome-mediated proteolysis than plasma membrane-located TRPV1. We conclude that the mechanism by which BoNT-A reduces TRPV1 expression involves the inhibition of TRPV1 plasma membrane trafficking and proteasome-mediated degradation in the cytoplasm. This paradigm seems to explain how BoNT-A alleviates TRPV1-mediated pain. Our data reveal a likely molecular mechanism whereby BoNT-A treatment reduces TRPV1 expression in the trigeminal system and provide important clues to novel therapeutic measures for ameliorating craniofacial pain.


Botulinum Toxins, Type A/pharmacology , Neuromuscular Agents/pharmacology , Neurons/drug effects , Neurons/metabolism , TRPV Cation Channels/biosynthesis , Animals , Blotting, Western , Immunohistochemistry , Male , PC12 Cells , Pain/drug therapy , Pain/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism
17.
Neuroreport ; 23(9): 560-5, 2012 Jun 20.
Article En | MEDLINE | ID: mdl-22562048

TRPV1 is a nonselective cation channel in nociceptors. TRPV1 stimulation has been shown to lead to the activation of microglia and astrocytes in the dorsal horn of the spinal cord. However, information on the effect of TRPV1 stimulation on glial activation in the trigeminal nucleus caudalis (TNC) is lacking. Here, we stimulated TRPV1 in the trigeminal afferents by a repetitive injection of 10 mmol/l capsaicin into the whisker pad for 2 days (d2 group), 4 days (d4 group), or 6 days (d6 group). As a control (c group), the vehicle was injected for 2 days. Anti-Iba1 and anti-glial fibrillary acidic protein antibodies were used to immunostain microglia and astrocytes in the TNC, respectively. The ratio of the cross-sectional area immunoreactive for Iba1 to the entire area of the TNC was increased in the d2 group compared with the c group on the injected side. Microglia were recruited to the superficial layers of the TNC. The numbers of microglia were reduced in the d4 group and the d6 group compared with the d2 group. The ratio of the cross-sectional area immunoreactive for glial fibrillary acidic protein to the entire TNC showed a significant increase in d2 group and the d4 group compared with the c group on the injected side. Behavioral analysis indicated that mechanical allodynia began to develop after 2 days of capsaicin treatment and persisted for at least 6 days after the onset of the repetitive capsaicin injection. These data indicate that TRPV1 stimulation activates the microglia and astrocytes in temporally distinct ways and that the development of mechanical allodynia is independent of such glial activation.


Astrocytes/metabolism , Hyperalgesia/metabolism , Microglia/metabolism , Nociceptors/metabolism , TRPV Cation Channels/agonists , Trigeminal Caudal Nucleus/metabolism , Animals , Astrocytes/drug effects , Behavior, Animal/drug effects , Calcium-Binding Proteins/drug effects , Calcium-Binding Proteins/metabolism , Capsaicin/pharmacology , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Microfilament Proteins/drug effects , Microfilament Proteins/metabolism , Microglia/drug effects , Nociceptors/drug effects , Rats , Rats, Sprague-Dawley , Sensory System Agents/pharmacology , Trigeminal Caudal Nucleus/drug effects
18.
Haematologica ; 93(10): 1495-504, 2008 Oct.
Article En | MEDLINE | ID: mdl-18728031

BACKGROUND: Thrombocytopenia is a common problem in the management of patients with cancer and other conditions that affect hematopoietic cells. In previous clinical trials, the polyethylene-glycol-conjugated recombinant human megakaryocyte growth and development factor increased platelet counts in patients with idiopathic thrombocytopenic purpura and solid tumors undergoing chemotherapy. However, antibodies to polyethylene-glycol-conjugated recombinant human megakaryocyte growth and development factor develop in healthy volunteers and patients undergoing chemotherapy and cross-react with endogenous thrombopoietin. As a result, clinical development of polyethylene-glycol-conjugated recombinant human megakaryocyte growth and development factor was discontinued in 1998. The aim of this study was to identify an orally bioavailable human Mpl activator that does not develop autoantibodies against endogenous thrombopoietin. DESIGN AND METHODS: We screened our chemical library and created a novel non-peptidyl thrombopoietin receptor, Mpl activator named butyzamide. We evaluated the effect of butyzamide on megakaryopoiesis in vitro using Ba/F3 cells expressing Mpl and human hematopoietic stem cells. For the evaluation of its in vivo effect, we administered butyzamide orally to immunodeficient NOD/Shi-scid,IL-2R gamma(null) (NOG) mice transplanted with human fetal liver-derived CD34(+) cells and investigated the production of human platelets. RESULTS: Butyzamide specifically reacted with human Mpl and activated the same signal transduction pathway as thrombopoietin. However, unlike thrombopoietin, butyzamide did not react with murine Mpl and was shown to require the histidine residue in the transmembrane domain of Mpl for its agonistic activity. Butyzamide induced colony-forming unit-megakaryocytes and polyploid megakaryocytes from human CD34(+) hematopoietic progenitor cells, and its effects were comparable to those of thrombopoietin. When butyzamide was administered orally at the doses of 10 and 50 mg/kg for 20 days to NOG mice transplanted with human fetal liver-derived CD34(+) cells, the human platelet count increased by 6.2- and 22.9-fold, respectively. CONCLUSIONS: Butyzamide is an orally bioavailable human Mpl activator, and appears to have potential for clinical development as a therapeutic agent for patients with thrombocytopenia.


Cell Differentiation/drug effects , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Receptors, Thrombopoietin/metabolism , Thiazoles/pharmacology , Animals , Antigens, CD34/metabolism , Blood Platelets/cytology , Blood Platelets/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Megakaryocytes/cytology , Methacrylates , Mice , Molecular Structure , Peptides/pharmacology , Platelet Aggregation/drug effects , Receptors, Thrombopoietin/genetics , Signal Transduction/drug effects , Thiazoles/chemistry , Thrombopoietin/agonists , Thrombopoietin/metabolism
19.
Biosci Rep ; 28(5): 275-85, 2008 Oct.
Article En | MEDLINE | ID: mdl-18620546

We have identified a series of novel non-peptide compounds that activate the thrombopoietin-dependent cell line Ba/F3-huMPL. The compounds stimulated proliferation of Ba/F3-huMPL in the absence of other growth factors, but did not promote proliferation of the thrombopoietin-independent parent cell line Ba/F3. The thrombopoietin-mimetic compounds elicited signal-transduction responses comparable with recombinant human thrombopoietin, such as tyrosine phosphorylation of the thrombopoietin receptor, JAK (Janus kinase) 2, Tyk2 (tyrosine kinase 2), STAT (signal transducer and activator of transcription) 3, STAT5, MAPKs (mitogen-activated protein kinases), PLCgamma (phospholipase Cgamma), Grb2 (growth-factor-receptor-bound protein 2), Shc (Src homology and collagen homology), Vav, Cbl and SHP-2 (Src homology 2 domain-containing protein tyrosine phosphatase 2) and increased the number of CD41(+) cells (megakaryocyte lineage) in cultures of human CD34(+) bone-marrow cells (haematopoietic stem cells). These findings suggest that this series of compounds are novel agonists of the human thrombopoietin receptor and are possible lead compounds for the generation of anti-thrombocytopaenia drugs.


Biomimetic Materials/pharmacology , Bone Marrow Cells/metabolism , Receptors, Thrombopoietin/agonists , Signal Transduction/drug effects , Thrombopoiesis/drug effects , Thrombopoietin/pharmacology , Animals , Bone Marrow Cells/cytology , Cell Line , GRB2 Adaptor Protein/biosynthesis , Humans , Mice , Phospholipase C gamma/biosynthesis , Protein Kinases/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/biosynthesis , Proto-Oncogene Proteins c-cbl/biosynthesis , Proto-Oncogene Proteins c-vav/biosynthesis , Receptors, Thrombopoietin/metabolism , STAT3 Transcription Factor/biosynthesis , STAT5 Transcription Factor/biosynthesis , Shc Signaling Adaptor Proteins/biosynthesis
...