Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 146
1.
Mol Neurobiol ; 58(11): 5602-5617, 2021 Nov.
Article En | MEDLINE | ID: mdl-34368932

Subarachnoid hemorrhage (SAH) is a catastrophic form of stroke responsible for significant morbidity and mortality. Oxidative stress, inflammation, and neuronal apoptosis are important in the pathogenesis of early brain injury (EBI) following SAH. Preconditioning exercise confers neuroprotective effects, mitigating EBI; however, the basis for such protection is unknown. We investigated the effects of preconditioning exercise on brain damage and sensorimotor function after SAH. Male rats were assigned to either a sham-operated (Sham) group, exercise (Ex) group, or no-exercise (No-Ex) group. After a 3-week exercise program, they underwent SAH by endovascular perforation. Consciousness level, neurological score, and sensorimotor function were studied. The expression of nuclear factor erythroid 2 p45-related factor 2 (Nrf2), heme oxygenase 1 (HO-1), 4-hydroxynonenal (4HNE), nitrotyrosine (NT), ionized calcium-binding adaptor molecule 1 (Iba1), tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), interleukin 1ß (IL-1ß), 14-3-3γ, p-ß-catenin Ser37, Bax, and caspase-3 were evaluated by immunohistochemistry or western blotting. The terminal deoxynucleotidyl transferase-mediated biotinylated dUTP nick end labeling (TUNEL) assay was also performed. After SAH, the Ex group had significantly reduced neurological deficits, sensorimotor dysfunction, and consciousness disorder compared with the No-Ex group. Nrf2, HO-1, and 14-3-3γ were significantly higher in the Ex group, while 4HNE, NT, Iba1, TNF-α, IL-6, IL-1ß, Bax, caspase-3, and TUNEL-positive cells were significantly lower. Our findings suggest that preconditioning exercise ameliorates EBI after SAH. The expression of 4HNE and NT was reduced by Nrf2/HO-1 pathway activation; additionally, both oxidative stress and inflammation were reduced. Furthermore, preconditioning exercise reduced apoptosis, likely via the 14-3-3γ/p-ß-catenin Ser37/Bax/caspase-3 pathway.


Brain Damage, Chronic/prevention & control , Neurons/pathology , Physical Conditioning, Animal , Subarachnoid Hemorrhage/complications , 14-3-3 Proteins/physiology , Animals , Apoptosis , Brain Damage, Chronic/diagnostic imaging , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation , Image Processing, Computer-Assisted , In Situ Nick-End Labeling , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/prevention & control , Oxidative Stress , Physical Conditioning, Animal/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors , X-Ray Microtomography
2.
Neuromolecular Med ; 23(1): 211-223, 2021 03.
Article En | MEDLINE | ID: mdl-32914259

Sphingosine 1-phosphate (S1P) is an important lipid biomolecule that exerts pleiotropic cellular actions as it binds to and activates its five G-protein-coupled receptors, S1P1-5. Through these receptors, S1P can mediate diverse biological activities in both healthy and diseased conditions. S1P is produced by S1P-producing enzymes, sphingosine kinases (SphK1 and SphK2), and is abundantly present in different organs, including the brain. The medically important roles of receptor-mediated S1P signaling are well characterized in multiple sclerosis because FTY720 (Gilenya™, Novartis), a non-selective S1P receptor modulator, is currently used as a treatment for this disease. In cerebral ischemia, its role is also notable because of FTY720's efficacy in both rodent models and human patients with cerebral ischemia. In particular, some of the S1P receptors, including S1P1, S1P2, and S1P3, have been identified as pathogenic players in cerebral ischemia. Other than these receptors, S1P itself and S1P-producing enzymes have been shown to play certain roles in cerebral ischemia. This review aims to compile the current updates and overviews about the roles of S1P signaling, along with a focus on S1P receptors in cerebral ischemia, based on recent studies that used in vivo rodent models of cerebral ischemia.


Brain Ischemia/metabolism , Lysophospholipids/physiology , Nerve Tissue Proteins/physiology , Sphingosine-1-Phosphate Receptors/physiology , Sphingosine/analogs & derivatives , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Brain Ischemia/complications , Clinical Trials as Topic , Disease Models, Animal , Drug Evaluation, Preclinical , Fingolimod Hydrochloride/therapeutic use , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Inflammation , Ischemic Stroke/drug therapy , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/physiology , Rats , Signal Transduction/physiology , Sphingosine/physiology
3.
Medicina (Kaunas) ; 56(3)2020 Mar 19.
Article En | MEDLINE | ID: mdl-32204311

Traumatic brain injury represents physical damage to the brain tissue that induces transitory or permanent neurological disabilities. The traumatic injury activates an important inflammatory response, followed by a cascade of events that lead to neuronal loss and further brain damage. Maintaining proper ventilation, a normal level of oxygenation, and adequate blood pressure are the main therapeutic strategies performed after injury. Surgery is often necessary for patients with more serious injuries. However, to date, there are no therapies that completely resolve the brain damage suffered following the trauma. Stem cells, due to their capacity to differentiate into neuronal cells and through releasing neurotrophic factors, seem to be a valid strategy to use in the treatment of traumatic brain injury. The purpose of this review is to provide an overview of clinical trials, aimed to evaluate the use of stem cell-based therapy in traumatic brain injury. These studies aim to assess the safety and efficacy of stem cells in this disease. The results available so far are few; therefore, future studies need in order to evaluate the safety and efficacy of stem cell transplantation in traumatic brain injury.


Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/therapy , Inflammation/etiology , Stem Cell Transplantation/methods , Adolescent , Adult , Aged , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Brain Damage, Chronic/therapy , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Child , Child, Preschool , Clinical Trials as Topic , Female , Humans , Male , Middle Aged , Nerve Growth Factors/metabolism , Neurons/pathology , Safety , Treatment Outcome , Young Adult
4.
Pharmacol Res ; 144: 331-335, 2019 06.
Article En | MEDLINE | ID: mdl-31042564

Dickkopf-1 (DKK1), a secretory glycoprotein discovered for 'inducing generation of head', is an endogenous inhibitor of the canonical Wnt/ß-catenin signaling pathway. It was found to be involved in many pathophysiological processes in vivo. Abnormal expression of DKK1 will alter expressions of related proteins and genes not only in canonical Wnt/ß-catenin signaling pathway but also in other signaling pathways. Previous studies of DKK1 focused on its function in tumors. In recent years, a large number of studies have shown that it plays an important role in embryonic development, neural regeneration, synaptogenesis and so on. Therefore, its role in neuropsychiatric disorders, such as neurodysplasia, cognitive impairment and emotional disorder, has attracted increasing attention. At present, the role of DKK1 in Alzheimer's disease (AD) is one of the research hot topics. This article reviewed the research progress of its role in AD in order to provide new ideas and directions for further studies on the pathogenesis and treatment of AD.


Alzheimer Disease/complications , Brain Damage, Chronic/etiology , Brain/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/pathology , Humans , Intercellular Signaling Peptides and Proteins/analysis , Wnt Signaling Pathway
5.
Am J Forensic Med Pathol ; 39(1): 8-13, 2018 Mar.
Article En | MEDLINE | ID: mdl-29293100

The polysialylated isoform of the neural cell adhesion molecule (PSA-NCAM) has been shown to be a key player in neuroplastic changes and is expressed in various disorders. We investigated the PSA-NCAM expression on brain cortical tissue in a cohort of drug-related deaths. Brains from 25 drug abusers and 10 control subjects were removed at autopsy, and 2 samples of the right parietal lobe of each case were obtained. The polysialylated isoform of NCAM was evaluated on formalin-fixed and paraffin-embedded tissues. Eleven patients were polydrug abusers; 14 used a single substance. The mechanisms of death were acute respiratory failure (n = 19), cardiorespiratory failure (n = 4), acute heart failure (n = 1), and brain injury (n = 1). Toxicological analyses of blood were available for all cases, and urine and bile analyses for 19 of 25 cases. The polysialylated isoform of NCAM immunoexpression in the neuronal soma and dendritic spines was observed in 18 (72%) of 25 drug abusers and in 2 (20%) of 10 control subjects. Drug abusers were statistically more positive for PSA-NCAM than control subjects (P = 0.0082). The expression of PSA-NCAM in the parietal cortex could be an indicator of brain damage due to drug abuse, and its availability could allow the forensic pathologists to develop rapid and low-cost additional or alternative method to improve detection of drug-related deaths.


Neural Cell Adhesion Molecule L1/metabolism , Parietal Lobe/metabolism , Sialic Acids/metabolism , Substance-Related Disorders/diagnosis , Adult , Biomarkers/metabolism , Brain Damage, Chronic/diagnosis , Brain Damage, Chronic/metabolism , Case-Control Studies , Female , Forensic Pathology , Humans , Immunohistochemistry , Male , Middle Aged , Protein Isoforms/metabolism , Substance-Related Disorders/metabolism , Young Adult
6.
Neuromolecular Med ; 19(4): 541-554, 2017 Dec.
Article En | MEDLINE | ID: mdl-28916896

The pro-inflammatory activity of interleukin 17, which is produced by the IL-23/IL-17 axis, has been associated with the pathogenesis of traumatic brain injury (TBI). The study investigated the potential role of IL-17 in secondary brain injury of TBI in a rat model. Our data showed that the levels of IL-17 increased from 6 h to 7 days and peaked at 3 days, in both the CNS and serum, which were consistent with the severity of secondary brain injury. The IL-23 inhibitor suberoylanilide hydroxamic acid (SAHA) treatment markedly decreased the expressions of IL-17 and apoptosis-associated proteins cleaved caspase-3 and increased the protein ratio of Bcl-2 (B cell lymphoma/leukemia-2)/Bax (Bcl-2-associated X protein). Meanwhile, neuronal apoptosis was reduced, and neural function was improved after SAHA treatment. This study suggests that IL-17 is involved in secondary brain injury after TBI. Administering an IL-23 inhibitor and thereby blocking the IL-23/IL-17 axis may be beneficial in the treatment of TBI.


Brain Damage, Chronic/physiopathology , Brain Injuries, Traumatic/physiopathology , Interleukin-17/physiology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/prevention & control , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Down-Regulation/drug effects , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Inflammation , Interleukin-17/blood , Interleukin-17/cerebrospinal fluid , Interleukin-17/genetics , Interleukin-23/antagonists & inhibitors , Interleukin-23/physiology , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/cerebrospinal fluid , Nerve Tissue Proteins/genetics , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Time Factors , Vorinostat
7.
Rev. neurol. (Ed. impr.) ; 61(9): 387-394, 1 nov., 2015. tab, graf
Article Es | IBECS | ID: ibc-145392

Introducción. Las alteraciones cognitivas, emocionales y comportamentales secundarias al daño cerebral adquirido y las demencias degenerativas pueden valorarse cuantitativa y cualitativamente mediante la administración de autoinformes que interroguen –a pacientes e informadores fiables– sobre las dificultades de los pacientes en la vida cotidiana. Sujetos y métodos. Se administró el inventario de síntomas prefrontales (ISP) y el cuestionario de fallos de memoria en la vida cotidiana modificado (MFE-30) a 174 participantes emparejados: 87 pacientes con daño cerebral o demencias degenerativas y sus 87 informadores fiables. Se exploró, junto con la bondad psicométrica de las pruebas, la utilidad clínica de la aplicación de estos cuestionarios a pacientes e informadores para obtener un índice de discrepancia de las puntuaciones como medida de la anosognosia. Resultados. Los resultados muestran cómo aplicar el ISP-20 (20 ítems) o el ISP (46 ítems), sean o no administrados conjuntamente con el MFE-30 (30 ítems), resulta un procedimiento muy útil para la valoración de la sintomatología en los individuos con daño cerebral adquirido o demencias degenerativas, al proporcionar una gran cantidad de información sobre las dificultades de los pacientes en la vida cotidiana. Conclusiones. Se recomienda, junto con la obligada evaluación neuropsicológica, la cumplimentación de cuestionarios o inventarios de síntomas como los propuestos, dado que presentan ventajas desde el punto de vista clínico, además de resultar eficaces, efectivos y eficientes en términos económicos (AU)


Introduction. The cognitive, emotional and behavioural alterations secondary to acquired brain injury and degenerative dementias can be quantitatively and quantitatively appraised by administering self-reports that ask both patients and reliable informants about the difficulties patients have in their everyday life. Subjects and methods. The Prefrontal Symptoms Inventory (PSI) and the Modified Memory Failures in Everyday Life Questionnaire (MFE-30) were administered to 174 paired participants: 87 patients with brain damage or degenerative dementias and their 87 reliable informants. In addition to the psychometric goodness of the tests, the study also explored the clinical usefulness of applying these questionnaires to patients and their informants in order to obtain a rate of discrepancy in the scores as a measure of anosognosia. Results. The results show how applying the PSI-20 (20 items) or the PSI (46 items), whether administered together with the MFE-30 (30 items) or not, is a very useful procedure for assessing the symptoms in individuals with acquired brain injury or degenerative dementias, since it yields a great deal of information about patients’ difficulties in their daily life. Conclusions. We recommend that, in addition to the compulsory neuropsychological assessment, questionnaires or inventories of symptoms like those proposed here should be carried out, due to the fact that they offer a number of advantages from the clinical point of view, as well as being efficacious and effective in economic terms (AU)


Female , Humans , Male , Neuropsychology/education , Neuropsychology/ethics , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/pathology , Dementia/psychology , Metabolism, Inborn Errors/genetics , Prefrontal Cortex/abnormalities , Surveys and Questionnaires/classification , Clinical Clerkship/methods , Mass Screening/analysis , Neuropsychology/methods , Neuropsychology/standards , Brain Damage, Chronic/complications , Brain Damage, Chronic/diagnosis , Dementia/pathology , Metabolism, Inborn Errors/metabolism , Prefrontal Cortex/injuries , Surveys and Questionnaires , Clinical Clerkship , Mass Screening/methods
8.
Rev. neurol. (Ed. impr.) ; 60(6): 249-256, 16 mar., 2015. graf, tab
Article Es | IBECS | ID: ibc-134595

Introducción. Un 30-40% de los pacientes con daño cerebral presenta alteraciones del nivel de conciencia, y algunos casos, estados alterados de conciencia: síndrome de vigilia sin respuesta (SVSR) o estado de mínima conciencia (EMC). La recuperación es variable y la supervivencia está amenazada por múltiples complicaciones. Objetivos. Presentar la metodología de trabajo del Hospital La Pedrera (HLP) para pacientes en SVSR o EMC y analizar las características clínicas de los pacientes atendidos, la evolución, y la situación funcional y cognitiva en el momento del alta. Pacientes y métodos. Estudio descriptivo prospectivo de pacientes atendidos en el HLP durante el período 2009-2013, con diagnóstico de SVSR o EMC. Resultados. El HLP trabaja mediante el método gestión de caso, ofreciendo una atención integral por un equipo multidisciplinar. Los pacientes se clasifican según objetivos asistenciales. Los pacientes con SVSR o EMC se incluyen en el programa de cuidados integrales y adaptación. Se atendió a 23 pacientes (86,9% varones), con una edad media de 54,9 años. Etiología: hemorragia cerebral, 30,4%; encefalopatía anóxica, 26,6%; encefalopatía metabólica, 17,3%; y otras causas, 17,3%. El 73,9% ingresó en SVSR y el resto en EMC. Evolución: el 43,4% mejoró su situación cognitiva inicial y el 88,8% presentaba una situación de dependencia total en el momento del alta. Las complicaciones más frecuentes fueron infecciones respiratorias y urinarias (53,6%). El 65,2% de los casos fueron exitus. Conclusiones. La asistencia en SVSR o EMC es compleja y precisa cuidados multidisciplinares. Casi la mitad de los pacientes mejoró su situación cognitiva, lo que justifica una actitud proactiva que intente mejorar la calidad de vida de los pacientes y sus familias (AU)


Introduction. Between 30% and 40% of patients with brain damage present alterations in their level of consciousness and, in some cases, altered states of consciousness: unresponsive wakefulness syndrome (UWS) or minimally conscious state (MCS). Recovery varies and survival is threatened by a number of complications. Aims. The purpose of this study is to present the working methodology used at the Hospital La Pedrera (HLP) for patients in UWS or MCS and to analyse the clinical characteristics of the patients attended to, their progress, and the functional and cognitive situation at the time of their discharge from hospital. Patients and methods. The work consisted in a prospective descriptive study of patients seen at the HLP over the period 2009-2013, who had been diagnosed with UWS or MCS. Results. The HLP uses the case management method, offering integrated care dispensed by a multidisciplinary team. Patients are classified according to healthcare goals. Patients with UWS or MCS are included in the integrated care and adaptation programme. A total of 23 patients (86.9% males) were attended to, the mean age being 54.9 years. Aetiology: brain haemorrhage, 30.4%; anoxic encephalopathy, 26.6%; metabolic encephalopathy, 17.3%; and other causes, 17.3%. Altogether 73.9% were admitted in UWS and the rest in MCS. Course: 43.4% improved their initial cognitive situation and 88.8% presented a situation of total dependence at the time of discharge. The most frequent complications were respiratory and urinary infections (53.6%). Death occurred in 65.2% of cases. Conclusions. Medical attention in UWS or MCS is complex and requires multidisciplinary care. Almost half of the patients improved their cognitive situation, which justifies a proactive attitude that attempts to improve the quality of life of both patients and their families (AU)


Humans , Male , Female , Brain Damage, Chronic/cerebrospinal fluid , Brain Damage, Chronic/metabolism , Consciousness Disorders/complications , Consciousness Disorders/pathology , Respiratory Tract Infections/diagnosis , Pharmaceutical Preparations/administration & dosage , Brain Damage, Chronic/complications , Brain Damage, Chronic/pathology , Consciousness Disorders/metabolism , Consciousness Disorders/prevention & control , Respiratory Tract Infections/metabolism , Pharmaceutical Preparations , Epidemiology, Descriptive
9.
J Nutr ; 144(7): 1058-66, 2014 Jul.
Article En | MEDLINE | ID: mdl-24744313

Gestational iron deficiency (ID) has been associated with a wide variety of central nervous system (CNS) impairments in developing offspring. However, a focus on singular regions has impeded an understanding of the CNS-wide effects of this micronutrient deficiency. Because the developing brain requires iron during specific phases of growth in a region-specific manner, we hypothesized that maternal iron deprivation would lead to region-specific impairments in the CNS of offspring. Female rats were fed an iron control (Fe+) or iron-deficient (Fe-) diet containing 240 or 6 µg/g iron during gestation and lactation. The corpus callosum (CC), hippocampus, and cortex of the offspring were analyzed at postnatal day 21 (P21) and/or P40 using structural and functional measures. In the CC at P40, ID was associated with reduced peak amplitudes of compound action potentials specific to myelinated axons, in which diameters were reduced by ∼20% compared with Fe+ controls. In the hippocampus, ID was associated with a 25% reduction in basal dendritic length of pyramidal neurons at P21, whereas branching complexity was unaffected. We also identified a shift toward increased proximal branching of apical dendrites in ID without an effect on overall length compared with Fe+ controls. ID also affected cortical neurons, but unlike the hippocampus, both apical and basal dendrites displayed a uniform decrease in branching complexity, with no significant effect on overall length. These deficits culminated in significantly poorer performance of P40 Fe- offspring in the novel object recognition task. Collectively, these results demonstrate that non-anemic gestational ID has a significant and region-specific impact on neuronal development and may provide a framework for understanding and recognizing the presentation of clinical symptoms of ID.


Brain Damage, Chronic/etiology , Cerebral Cortex/diagnostic imaging , Corpus Callosum/diagnostic imaging , Iron Deficiencies , Lactation , Maternal Nutritional Physiological Phenomena , Neurons/diagnostic imaging , Animals , Axons/metabolism , Axons/ultrastructure , Brain Damage, Chronic/congenital , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/pathology , Cerebral Cortex/metabolism , Corpus Callosum/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , Female , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Male , Nerve Fibers, Myelinated/diagnostic imaging , Nerve Fibers, Myelinated/metabolism , Neurogenesis , Neurons/metabolism , Pregnancy , Pyramidal Cells/diagnostic imaging , Pyramidal Cells/metabolism , Random Allocation , Rats , Rats, Inbred F344 , Reproducibility of Results , Ultrasonography
10.
Biol Trace Elem Res ; 158(2): 176-85, 2014 May.
Article En | MEDLINE | ID: mdl-24573406

Selenium (Se) is an important dietary micronutrient with antioxidative roles. Cadmium (Cd), a ubiquitous environmental pollutant, is known to cause brain lesion in rats and humans. However, little is reported about the deleterious effects of subchronic Cd exposure on the brain of poultry and the protective roles on the brain by Se against Cd. The aim of this study was to investigate the protective effects of Se on Cd-induced brain damage in chickens. One hundred twenty 100-day-old chickens were randomly assigned to four groups and were fed a basal diet, or Se (as 10 mg Na2SeO3/kg dry weight of feed), Cd (as 150 mg CdCl2/kg dry weight of feed), or Cd + Se in their basic diets for 60 days. Then, concentrations of Cd and Se, production of nitric oxide (NO), messenger RNA (mRNA) level and activity of inducible NO synthase (iNOS), level of oxidative stress, and histological and ultrastructural changes of the cerebrum and cerebellum were examined. The results showed that Cd exposure significantly increased Cd accumulation, NO production, iNOS activities, iNOS mRNA level, and MDA content in the cerebrum and cerebellum. Cd treatment obviously decreased Se content and antioxidase activities and caused histopathological changes in the cerebrum and cerebellum. Se supplementation during dietary Cd obviously reduced Cd accumulation, NO production, mRNA level and activity of iNOS, oxidative stress, and histopathological damage in the cerebrum and cerebellum of chickens. It indicated that Se ameliorates Cd-induced brain damage in chickens by regulating iNOS-NO system changes, and oxidative stress induced by Cd and Se can serve as a potential therapeutic for Cd-induced brain lesion of chickens.


Brain Damage, Chronic/chemically induced , Brain Damage, Chronic/prevention & control , Cadmium/toxicity , Selenium/pharmacology , Animals , Brain/drug effects , Brain/enzymology , Brain/metabolism , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/metabolism , Cadmium/administration & dosage , Chickens , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Selenium/administration & dosage
11.
Resuscitation ; 85(2): 299-305, 2014 Feb.
Article En | MEDLINE | ID: mdl-24200890

OBJECTIVE: The aim of this study was to investigate whether the DOR agonist BW373U86 conferred neuroprotection following ACA when given after resuscitation and to determine the long-term effects of chronic BW373U86 treatment on ACA-elicited brain injury. METHODS: Animals were divided into acute and chronic treatment groups. Each group consisted of four sub-groups, including Sham, ACA, BW373U86 (BW373U86+ACA), and Naltrindole groups (Naltrindole and BW373U86+ACA). The DOR antagonist Naltrindole was used to confirm the possible receptor-dependent effects of BW373U86. ACA was induced by 8min of asphyxiation followed by resuscitation. All drugs were administered either immediately after the restoration of spontaneous circulation (ROSC) in acute-treatment groups or over 6 consecutive days in chronic-treatment groups. Alterations of cAMP response element-binding protein (CREB) and phosphorylated CREB (pCREB) were analyzed by western blot and immunohistochemistry. Neurological functions were assessed by neurological deficit score (NDS) and Morris Water Maze performance. Neurodegeneration was monitored by immunofluorescence and Nissl staining. RESULTS: ACA induced massive neuron loss and serious neurological function deficits. BW373U86 significantly reduced both of these negative effects and increased CREB and pCREB expression in the hippocampus; these effects were reversed with acute Naltrindole treatment. The protective effects of BW373U86 persisted until 28d post-ROSC with chronic treatment, but these effects were not reversed by Naltrindole. CONCLUSIONS: BW373U86 attenuates global cerebral ischemic injury induced by ACA through both DOR-dependent and DOR-independent mechanisms. CREB might be an important molecule in mediating these neuroprotective effects.


Asphyxia/therapy , Benzamides/pharmacology , Brain Damage, Chronic/prevention & control , Cardiopulmonary Resuscitation , Heart Arrest/therapy , Neuroprotective Agents/pharmacology , Piperazines/pharmacology , Animals , Blotting, Western , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/pathology , CREB-Binding Protein/metabolism , Disease Models, Animal , Immunoenzyme Techniques , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Rats, Sprague-Dawley
12.
Brain Res ; 1455: 114-23, 2012 May 21.
Article En | MEDLINE | ID: mdl-22483792

Excess activation of ionotropic glutamate receptors and iron is believed to contribute to free radical production and neuronal death following hypoxic ischemia. We examined the possibility that both NMDA receptor activation and iron overload determine spatial and temporal patterns of free radical production after transient middle cerebral artery occlusion (tMCAO) in male Sprague-Dawley rats. Mitochondrial free radical (MFR) levels were maximally increased in neurons in the core at 1 h and 24 h after tMCAO. Early MFR production was blocked by administration of MK-801, an NMDA receptor antagonist, but not deferoxamine, an iron chelator. Neither MK-801 nor deferoxamine attenuated late MFR production in the core. Increased MFRs were observed in penumbral neurons within 6 h and gradually increased over 24 h after tMCAO. Slowly-evolving MFRs in the core and penumbra were accompanied by iron overload. Deferoxamine blocked iron overload but reduced MFR production only in the penumbra. Combined MK-801/deferoxamine reduced late MFR production in both core and penumbra in an additive manner. Combination therapy significantly ameliorated infarction compared with monotherapy. These findings suggest that the NMDA receptor activation and iron overload mediate late MFR production and infarction after tMCAO.


Brain Damage, Chronic/metabolism , Free Radicals/metabolism , Infarction, Middle Cerebral Artery/metabolism , Iron/metabolism , Oxidative Stress/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Brain Damage, Chronic/drug therapy , Brain Damage, Chronic/physiopathology , Deferoxamine/pharmacology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
13.
Eur J Pharmacol ; 680(1-3): 41-8, 2012 Apr 05.
Article En | MEDLINE | ID: mdl-22314218

Chronic cerebral hypoperfusion, induced by permanent occlusion of bilateral common carotid arteries (2VO), is related to neurological disorders and contributes to cognitive decline. Chrysin (5,7-dihydroxyflavone) is an important member of the flavonoid family. The aim of this study is to investigate the effects of chrysin on cognitive deficits and brain damage in this rat 2VO model. At 52days after ligation, the escape latency in Morris water maze was significantly increased in rats subjected to 2VO, the neuronal damage was also increased accompanied by a large proliferation in glial fibrillary acidic protein (GFAP) immunoreactivity with marked white matter lesions, and neuronal cell apoptosis, all of which were significantly alleviated by long treatment of chrysin (30mg/kg). Biochemical examinations revealed that chrysin decreased lipid peroxide, reduced the increased activities of superoxide dismutase, and attenuated the decreased activities of glutathione peroxidase in 2VO rats. The results suggest that chrysin may have therapeutic potential for the treatment of neurodegeneration and dementia caused by decreased cerebral blood flow, which is most likely related, at least in part, to its anti-inflammatory and antioxidant properties.


Brain Damage, Chronic/drug therapy , Brain Ischemia/complications , Cognition Disorders/drug therapy , Flavonoids/pharmacology , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Cerebrovascular Circulation/drug effects , Cognition Disorders/metabolism , Cognition Disorders/pathology , Dementia/drug therapy , Dementia/metabolism , Dementia/pathology , Glial Fibrillary Acidic Protein/metabolism , Glutathione Peroxidase/metabolism , Learning Disabilities/drug therapy , Learning Disabilities/metabolism , Learning Disabilities/pathology , Lipid Peroxides/metabolism , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/metabolism , Memory Disorders/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
14.
Brain Pathol ; 22(2): 142-9, 2012 Mar.
Article En | MEDLINE | ID: mdl-21714827

While a history of a single traumatic brain injury (TBI) is associated with the later development of syndromes of cognitive impairment such as Alzheimer's disease, the long-term pathology evolving after single TBI is poorly understood. However, a progressive tauopathy, chronic traumatic encephalopathy, is described in selected cohorts with a history of repetitive concussive/mild head injury. Here, post-mortem brains from long-term survivors of just a single TBI (1-47 years survival; n=39) vs. uninjured, age-matched controls (n=47) were examined for neurofibrillary tangles (NFTs) and amyloid-ß (Aß) plaques using immunohistochemistry and thioflavine-S staining. Detailed maps of findings permitted classification of pathology using semiquantitative scoring systems. NFTs were exceptionally rare in young, uninjured controls, yet were abundant and widely distributed in approximately one-third of TBI cases. In addition, Aß-plaques were found in a greater density following TBI vs. controls. Moreover, thioflavine-S staining revealed that while all plaque-positive control cases displayed predominantly diffuse plaques, 64% of plaque-positive TBI cases displayed predominantly thioflavine-S-positive plaques or a mixed thioflavine-S-positive/diffuse pattern. These data demonstrate that widespread NFT and Aß plaque pathologies are present in up to a third of patients following survival of a year or more from a single TBI. This suggests that a single TBI induces long-term neuropathological changes akin to those found in neurodegenerative disease.


Amyloid beta-Peptides/adverse effects , Brain Damage, Chronic/pathology , Brain Injuries/pathology , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , tau Proteins/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Case-Control Studies , Female , Humans , Male , Middle Aged , Young Adult
15.
Glia ; 59(12): 1958-73, 2011 Dec.
Article En | MEDLINE | ID: mdl-21956849

NG2-expressing cells comprise a population of cycling precursors that can exit the cell cycle and differentiate into mature oligodendrocytes. As a whole, they display heterogeneous properties and behaviors that remain unresolved at the molecular level, although partly interpretable as distinct maturation stages. To address this issue, we analyzed the expression of the GPR17 receptor, recently shown to decorate NG2-expressing cells and to operate as an early sensor of brain damage, in immature and adult oligodendrocyte progenitors in the intact brain and after injury. In both the early postnatal and adult cerebral cortex, distinct GPR17 protein localizations and expression levels define different stages of oligodendroglial maturation, ranging from the precursor phase to the premyelinating phenotype. As soon as cells exit mitosis, a fraction of NG2-expressing cells displays accumulation of GPR17 protein in the Golgi apparatus. GPR17 expression is subsequently upregulated and distributed to processes of cells that stop dividing, progressively lose NG2 positivity and assume premyelinating features. Absence of colabeling with mature markers or myelin proteins indicates that GPR17 is downregulated when cells complete their final maturation. BrdU-based fate-mapping demonstrated that a significant fraction of newly generated oligodendrocyte progenitors transiently upregulates GPR17 during maturation. Importantly, we also found that GPR17 does not participate to the early reaction of NG2-expressing cells to damage, while it is induced at postacute stages after injury. These findings identify GPR17 as a marker for progenitor progression within the oligodendroglial lineage and highlight its participation to postacute reactivity of NG2 cells in different injury paradigms.


Antigens/biosynthesis , Brain Damage, Chronic/metabolism , Brain Injuries/metabolism , Cell Differentiation/physiology , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Proteoglycans/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Acute Disease , Animals , Antigens/genetics , Biomarkers/metabolism , Brain Damage, Chronic/pathology , Brain Injuries/pathology , Disease Models, Animal , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Oligodendroglia/pathology , Primary Cell Culture , Proteoglycans/genetics , Stem Cells/pathology
16.
Hypertension ; 58(4): 635-42, 2011 Oct.
Article En | MEDLINE | ID: mdl-21859961

The role of the renin-angiotensin system in cognitive impairment is unclear. This work was undertaken to test our hypothesis that renin-angiotensin system may contribute to cognitive decline and brain damage caused by chronic cerebral ischemia. C57BL/6J mice were subjected to bilateral common carotid artery stenosis with microcoil to prepare mice with chronic cerebral hypoperfusion, a model of subcortical vascular dementia. The effects of aliskiren, a direct renin inhibitor, or Tempol, a superoxide scavenger, on brain damage and working memory in these mice were examined. Chronic cerebral hypoperfusion significantly increased brain renin activity and angiotensinogen expression in C57BL/6J mice, which was attributed to the increased renin in activated astrocytes and microvessels and the increased angiotensinogen in activated astrocytes in white matter. Aliskiren pretreatment significantly inhibited brain renin activity and ameliorated brain p67(phox)-related NADPH oxidase activity, oxidative stress, glial activation, white matter lesion, and spatial working memory deficits in C57BL/6J mice with bilateral common carotid artery stenosis. To elucidate the role of oxidative stress in brain protective effects of aliskiren, we also examined the effect of Tempol in the same mice with bilateral common carotid artery stenosis. Tempol pretreatment mimicked the brain protective effects of aliskiren in this mouse model. Posttreatment of mice with aliskiren or Tempol after bilateral common carotid artery stenosis also prevented cognitive decline. In conclusion, chronic cerebral hypoperfusion induced the activation of the brain renin-angiotensin system. Aliskiren ameliorated brain damage and working memory deficits in the model of chronic cerebral ischemia through the attenuation of oxidative stress. Thus, direct renin inhibition seems to be a promising therapeutic strategy for subcortical vascular dementia.


Brain Damage, Chronic/prevention & control , Brain Ischemia/metabolism , Cognition Disorders/prevention & control , Dementia, Vascular/metabolism , Renin/antagonists & inhibitors , Renin/metabolism , Amides/pharmacology , Angiotensinogen/metabolism , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Brain Damage, Chronic/epidemiology , Brain Damage, Chronic/metabolism , Cognition Disorders/epidemiology , Cognition Disorders/metabolism , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Fumarates/pharmacology , Hypertension/complications , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Risk Factors , Spin Labels
17.
J Neurotrauma ; 28(6): 983-93, 2011 Jun.
Article En | MEDLINE | ID: mdl-21501066

Inflammatory and ischemic processes contribute to the development of secondary brain damage after mechanical brain injury. Recent data suggest that thiazolidinediones (TZDs), a class of drugs approved for the treatment of non-insulin-dependent diabetes mellitus, effectively reduces inflammation and brain lesion by stimulation of the peroxisome proliferator-activated receptor-γ (PPAR-γ). The present study investigates the influence of the TZD pioglitazone and rosiglitazone on inflammation and secondary brain damage after experimental traumatic brain injury (TBI). A controlled cortical impact (CCI) injury was induced in male C57BL/6 mice to investigate following endpoints: (1) mRNA expression of PPAR-γ and PPAR-γ target genes (LPL, GLT1, and IRAP/Lnpep), and inflammatory markers (TNF-α, IL-1ß, IL-6, and iNOS), at 15 min, 3 h, 6 h, 12 h, and 24 h post-trauma; (2) contusion volume, neurological function, and gene expression after 24 h in mice treated with pioglitazone (0.5 and 1 mg/kg) or rosiglitazone (5 and 10 mg/kg IP at 30 min post-trauma); and (3) the role of PPAR-γ to mediate protection was determined in animals treated with pioglitazone, the PPAR-γ inhibitor T0070907, and a combination of both. Inflammatory marker genes, but not PPAR-γ gene expression, was upregulated after trauma. Pioglitazone reduced the histological damage and inflammation in a dose-dependent fashion. In contrast, rosiglitazone failed to suppress inflammation and histological damage. PPAR-γ and PPAR-γ target gene expression was not induced by pioglitazone and rosiglitazone. In line with these results, pioglitazone-mediated protection was not reversed by T0070907. The results indicate that the neuroprotective effects of pioglitazone are not solely related to PPAR-γ-dependent mechanisms.


Brain Damage, Chronic/drug therapy , Brain Injuries/drug therapy , Hypoglycemic Agents/pharmacology , Neuroprotective Agents/pharmacology , PPAR gamma/physiology , Thiazolidinediones/pharmacology , Animals , Brain Damage, Chronic/metabolism , Brain Injuries/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Pioglitazone
18.
Brain Res ; 1382: 259-65, 2011 Mar 25.
Article En | MEDLINE | ID: mdl-21241675

Pristanic acid (Prist) is accumulated in various peroxisomal disorders characterized by severe neurological dysfunction whose pathogenesis is poorly understood. Since oxidative damage has been demonstrated in brain of patients affected by neurodegenerative disorders, in the present work we investigated the in vitro effects of Prist on important parameters of oxidative stress in cerebral cortex from young rats. Prist significantly increased malondialdehyde levels, reflecting an increase of lipid peroxidation. This effect was totally prevented by the free radical scavenger melatonin, suggesting the involvement of reactive species. Prist also provoked protein oxidative damage, as determined by increased carbonyl formation and sulfhydryl oxidation. Otherwise, it did not alter nitric oxide production, indicating that nitrogen reactive species were not implicated in the lipid and oxidative damage provoked by Prist. Furthermore, the concentration of glutathione (GSH), the major brain non-enzymatic antioxidant defense, was significantly decreased by Prist and this decrease was fully prevented by melatonin and attenuated by α-tocopherol. It is therefore presumed that Prist elicits oxidative stress in the brain probably via reactive oxygen species formation and that this pathomechanism may possibly be involved in the brain damage found in patients affected by peroxisomal disorders where Prist accumulates.


Brain Chemistry/drug effects , Brain Damage, Chronic/chemically induced , Cerebral Cortex/drug effects , Fatty Acids/toxicity , Oxidative Stress/drug effects , Peroxisomal Disorders/chemically induced , Animals , Brain Chemistry/physiology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Cerebral Cortex/growth & development , Cerebral Cortex/physiopathology , Disease Models, Animal , Fatty Acids/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Oxidative Stress/physiology , Peroxisomal Disorders/metabolism , Peroxisomal Disorders/physiopathology , Rats , Rats, Wistar
19.
Brain Res Rev ; 67(1-2): 157-83, 2011 Jun 24.
Article En | MEDLINE | ID: mdl-21238488

Almost a quarter of a century ago, interleukin-6 (IL-6) was discovered as an inflammatory cytokine involved in B cell differentiation. Today, IL-6 is recognized to be a highly versatile cytokine, with pleiotropic actions not only in immune cells, but also in other cell types, such as cells of the central nervous system (CNS). The first evidence implicating IL-6 in brain-related processes originated from its dysregulated expression in several neurological disorders such as multiple sclerosis, Alzheimer's disease and Parkinson's disease. In addition, IL-6 was shown to be involved in multiple physiological CNS processes such as neuron homeostasis, astrogliogenesis and neuronal differentiation. The molecular mechanisms underlying IL-6 functions in the brain have only recently started to emerge. In this review, an overview of the latest discoveries concerning the actions of IL-6 in the nervous system is provided. The central position of IL-6 in the neuroinflammatory reaction pattern, and more specifically, the role of IL-6 in specific neurodegenerative processes, which accompany Alzheimer's disease, multiple sclerosis and excitotoxicity, are discussed. It is evident that IL-6 has a dichotomic action in the CNS, displaying neurotrophic properties on the one hand, and detrimental actions on the other. This is in agreement with its central role in neuroinflammation, which evolved as a beneficial process, aimed at maintaining tissue homeostasis, but which can become malignant when exaggerated. In this perspective, it is not surprising that 'well-meant' actions of IL-6 are often causing harm instead of leading to recovery.


Interleukin-6/physiology , Mental Disorders/metabolism , Mental Disorders/pathology , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/pathology , Cell Death/physiology , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Interleukin-6/toxicity , Mental Disorders/etiology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
20.
Minerva Pediatr ; 62(3 Suppl 1): 141-3, 2010 Jun.
Article It | MEDLINE | ID: mdl-21090084

Hypoxia-ischemia (H-I) constitutes the main phenomenon responsible for brain-blood barrier permeability modifications leading to cerebral vascular autoregulation loss in newborns. Hypotension, cerebral ischemia, and reperfusion are the main events involved in vascular auto-regulation loss leading to cell death and tissue damage. Reperfusion could be critical since organ damage, particularly of the brain, may be amplified during this period. An exaggerated activation of vasoactive agents, of calcium mediated effects could be responsible for reperfusion injury (R-I), which, in turns, leads to cerebral hemorrhage and damage. These phenomena represent a common repertoire in newborns complicated by perinatal acute or chronic hypoxia treated by risky procedures such as mechanical ventilation, nitric oxide supplementation, brain cooling, and extracorporeal membrane oxygenation (ECMO). Despite accurate monitoring, the post-insult period is crucial, as clinical symptoms and standard monitoring parameters may be silent at a time when brain damage is already occurring and the therapeutic window for pharmacological intervention is limited. Therefore, the measurement of circulating biochemical markers of brain damage, such as vasoactive agents and nervous tissue peptides is eagerly awaited in clinical practice to detect high risk newborns. The present article is aimed at investigating the role of dosage biochemical markers in non-invasive biological fluids such as S100B, a calcium binding protein, activin A, a protein expressed in Central nervous System (CNS).


Activins/urine , Brain Damage, Chronic/prevention & control , Hypoxia-Ischemia, Brain/metabolism , Nerve Growth Factors/analysis , S100 Proteins/analysis , Saliva/chemistry , Biomarkers/analysis , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Case-Control Studies , Dimerization , Humans , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/physiopathology , Infant, Newborn , Infant, Newborn, Diseases/metabolism , Nerve Growth Factors/chemistry , Nerve Growth Factors/urine , Reperfusion Injury/prevention & control , S100 Calcium Binding Protein beta Subunit , S100 Proteins/chemistry , S100 Proteins/urine , Urinalysis
...