Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Medicina (Kaunas) ; 56(3)2020 Mar 19.
Article in English | MEDLINE | ID: mdl-32204311

ABSTRACT

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.


Subject(s)
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
2.
Pharmacol Res ; 144: 331-335, 2019 06.
Article in English | MEDLINE | ID: mdl-31042564

ABSTRACT

Dickkopf-1 (DKK1), a secretory glycoprotein discovered for 'inducing generation of head', is an endogenous inhibitor of the canonical Wnt/Ɵ-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/Ɵ-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.


Subject(s)
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
3.
Am J Forensic Med Pathol ; 39(1): 8-13, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29293100

ABSTRACT

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.


Subject(s)
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
4.
J Nutr ; 144(7): 1058-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24744313

ABSTRACT

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 Āµg/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 Ć¢ĀˆĀ¼20% 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.


Subject(s)
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
5.
Glia ; 59(12): 1958-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956849

ABSTRACT

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.


Subject(s)
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
6.
Mol Neurobiol ; 58(11): 5602-5617, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34368932

ABSTRACT

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 1Ɵ (IL-1Ɵ), 14-3-3ƎĀ³, p-Ɵ-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 14-3-3ƎĀ³ were significantly higher in the Ex group, while 4HNE, NT, Iba1, TNF-α, IL-6, IL-1Ɵ, 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 14-3-3ƎĀ³/p-Ɵ-catenin Ser37/Bax/caspase-3 pathway.


Subject(s)
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
7.
Neuromolecular Med ; 23(1): 211-223, 2021 03.
Article in English | MEDLINE | ID: mdl-32914259

ABSTRACT

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.


Subject(s)
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
8.
J Exp Med ; 189(4): 719-27, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9989987

ABSTRACT

The transcription factor interferon regulatory factor 1 (IRF-1) is involved in the molecular mechanisms of inflammation and apoptosis, processes that contribute to ischemic brain injury. In this study, the induction of IRF-1 in response to cerebral ischemia and its role in ischemic brain injury were investigated. IRF-1 gene expression was markedly upregulated within 12 h of occlusion of the middle cerebral artery in C57BL/6 mice. The expression reached a peak 4 d after ischemia (6.0 +/- 1.8-fold; P < 0.001) and was restricted to the ischemic regions of the brain. The volume of ischemic injury was reduced by 23 +/- 3% in IRF-1(+/-) and by 46 +/- 9% in IRF-1(-/-) mice (P < 0.05). The reduction in infarct volume was paralleled by a substantial attenuation in neurological deficits. Thus, IRF-1 is the first nuclear transacting factor demonstrated to contribute directly to cerebral ischemic damage and may be a novel therapeutic target in ischemic stroke.


Subject(s)
Brain Damage, Chronic/etiology , Brain Ischemia/metabolism , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation , Phosphoproteins/biosynthesis , Animals , Apoptosis/genetics , Brain Damage, Chronic/metabolism , Brain Ischemia/complications , Brain Ischemia/genetics , Brain Ischemia/pathology , Cerebral Infarction/pathology , DNA-Binding Proteins/genetics , Female , Genotype , Inflammation/genetics , Interferon Regulatory Factor-1 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Transcription, Genetic
9.
Metab Brain Dis ; 24(2): 283-98, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19294497

ABSTRACT

N-acetylaspartic acid (NAA) is the biochemical hallmark of Canavan Disease, an inherited metabolic disease caused by deficiency of aspartoacylase activity. NAA is an immediate precursor for the enzyme-mediated biosynthesis of N-acetylaspartylglutamic acid (NAAG), whose concentration is also increased in urine and cerebrospinal fluid of patients affected by CD. This neurodegenerative disorder is clinically characterized by severe mental retardation, hypotonia and macrocephaly, and generalized tonic and clonic type seizures. Considering that the mechanisms of brain damage in this disease remain not fully understood, in the present study we investigated whether intracerebroventricular administration of NAA or NAAG elicits oxidative stress in cerebral cortex of 30-day-old rats. NAA significantly reduced total radical-trapping antioxidant potential, catalase and glucose 6-phosphate dehydrogenase activities, whereas protein carbonyl content and superoxide dismutase activity were significantly enhanced. Lipid peroxidation indices and glutathione peroxidase activity were not affected by NAA. In contrast, NAAG did not alter any of the oxidative stress parameters tested. Our results indicate that intracerebroventricular administration of NAA impairs antioxidant defenses and induces oxidative damage to proteins, which could be involved in the neurotoxicity of NAA accumulation in CD patients.


Subject(s)
Aspartic Acid/analogs & derivatives , Canavan Disease/metabolism , Cerebral Cortex/metabolism , Neurotoxins/toxicity , Oxidative Stress/physiology , Animals , Antioxidants/metabolism , Aspartic Acid/administration & dosage , Aspartic Acid/metabolism , Aspartic Acid/toxicity , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Canavan Disease/complications , Catalase/drug effects , Catalase/metabolism , Cerebral Cortex/drug effects , Dipeptides/administration & dosage , Dipeptides/metabolism , Dipeptides/toxicity , Disease Models, Animal , Glucosephosphate Dehydrogenase/drug effects , Glucosephosphate Dehydrogenase/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Injections, Intraventricular , Lipid Peroxidation , Male , Neuropeptides/administration & dosage , Neuropeptides/metabolism , Neuropeptides/toxicity , Neurotoxins/administration & dosage , Neurotoxins/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Wistar
10.
Acta Neurochir (Wien) ; 151(11): 1419-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19499176

ABSTRACT

PURPOSE: Radiation-induced brain injury (RBI) is an insidious side-effect of radiotherapy mediated by vascular alterations, inflammation and ischaemia. In previous studies we had shown potential increases in loco-regional blood flow and glucose metabolism in brain tumours by using electrical cervical spinal cord stimulation (SCS). In this preliminary report we demonstrate the effect of cervical SCS on RBI-tissue metabolism, as assessed using [(18)F]fluorodeoxyglucose-positron emission tomography (FDG-PET). METHODS: SCS devices were inserted in eight patients with diagnosis of potential RBI in previously irradiated areas. While the SCS device was deactivated, each patient underwent an initial FDG-PET study to evaluate the clinical status. A second FDG-PET study was performed later the same day while the SCS device was activated in order to evaluate the effect of cervical SCS on glucose metabolism. RESULTS: Basal glucose metabolism in RBI areas was 31% lower than peri-RBI areas (p = 0.009) and 32% lower than healthy contra-lateral areas (p = 0.020). There was a significant increase in glucose uptake during SCS in both the RBI (p = 0.005) and the peri-RBI (p = 0.004) areas, with measured increases of 38 and 42%, respectively. The estimated potential maximal residual activity of the first FDG dose's contribution to the activity on the second scan was

Subject(s)
Brain Damage, Chronic/metabolism , Brain/metabolism , Glucose/metabolism , Radiation Injuries/metabolism , Radiotherapy/adverse effects , Spinal Cord/physiology , Adult , Afferent Pathways/physiology , Aged , Brain/diagnostic imaging , Brain/physiopathology , Brain Damage, Chronic/diagnostic imaging , Brain Damage, Chronic/physiopathology , Brain Mapping/methods , Brain Neoplasms/radiotherapy , Cerebrovascular Circulation/physiology , Cervical Vertebrae , Electric Stimulation/methods , Energy Metabolism/physiology , Female , Fluorodeoxyglucose F18 , Glucose/analysis , Humans , Male , Middle Aged , Positron-Emission Tomography/methods , Predictive Value of Tests , Radiation Injuries/diagnostic imaging , Radiation Injuries/physiopathology , Sensitivity and Specificity
11.
Circulation ; 115(12): 1599-608, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17372179

ABSTRACT

BACKGROUND: Stroke is the second to third leading cause of death. Toll-like receptor 4 (TLR4) is a signaling receptor in innate immunity that is a specific immunologic response to systemic bacterial infection and cerebral injury. The role of TLR4 in brain ischemia has not been examined yet. We have therefore investigated whether cerebral ischemia and inflammation produced by permanent occlusion of the middle cerebral artery differ in mice that lack a functional TLR4 signaling pathway. METHODS AND RESULTS: Permanent occlusion of the middle cerebral artery was performed on 2 strains of TLR4-deficient mice (C3H/HeJ and C57BL/10ScNJ) and respective controls (C3H/HeN and C57BL/10ScSn). Stroke outcome was evaluated by determination of infarct volume and assessment of neurological scores. Brains were collected 24 hours and 7 days after stroke. When compared with control mice, TLR4-deficient mice had lower infarct volumes and better outcomes in neurological and behavioral tests. Mice that lacked TLR4 had minor expression of stroke-induced interferon regulatory factor-1, inducible nitric oxide synthase, and cyclooxygenase-2, mediators implicated in brain damage. The levels of interferon-beta and of the lipid peroxidation marker malondialdehyde were also lower in brains from TLR4-deficient mice than in those from control mice. In addition, the expression of matrix metalloproteinase-9, which is induced and mediates brain damage, was also reduced in TLR4-deficient mice after experimental stroke. CONCLUSIONS: TLR4-deficient mice have minor infarctions and less inflammatory response after an ischemic insult. These data demonstrate that TLR4 signaling and innate immunity are involved in brain damage and in inflammation triggered by ischemic injury.


Subject(s)
Brain Damage, Chronic/pathology , Infarction, Middle Cerebral Artery/metabolism , Inflammation Mediators/metabolism , Toll-Like Receptor 4/physiology , Animals , Brain Chemistry , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/prevention & control , Cerebral Infarction/etiology , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/therapeutic use , Dinoprostone/analysis , Encephalitis/drug therapy , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/pathology , Gene Expression Regulation , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/genetics , Interferons/biosynthesis , Interferons/genetics , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Lipid Peroxidation , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitrobenzenes/therapeutic use , Oxidative Stress , Sulfonamides/therapeutic use , Toll-Like Receptor 4/deficiency , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
12.
Curr Opin Neurobiol ; 16(3): 258-64, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16713245

ABSTRACT

Brain insults cause rapid cell death, and a disruption of functional circuits, in the affected regions. As the injured tissue recovers from events associated with cell death, regenerative processes are activated that over months lead to a certain degree of functional recovery. Factors produced by new neurons and glia, axonal sprouting of surviving neurons, and new synapse formation help to re-establish some of the lost functions. The timing and location of such events is crucial in the success of the regenerative process. Comprehensive gene expression profiling and proteomic analyses have enabled a deeper molecular and cellular mechanistic understanding of post-injury brain regeneration. These new mechanistic insights are aiding the design of novel therapeutic modalities that enhance regeneration.


Subject(s)
Brain Damage, Chronic/physiopathology , Brain Injuries/physiopathology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Recovery of Function/physiology , Animals , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/therapy , Brain Injuries/metabolism , Brain Injuries/therapy , Cell Proliferation , Growth Cones/metabolism , Humans , Nerve Growth Factors/metabolism , Nerve Regeneration/drug effects , Neuronal Plasticity/drug effects , Recovery of Function/drug effects , Stem Cell Transplantation/trends
13.
Neurochem Int ; 52(6): 935-47, 2008 May.
Article in English | MEDLINE | ID: mdl-18093696

ABSTRACT

Epilepsy is a common neurological disorder that occurs more frequently in children than in adults. The extent that prolonged seizure activity, i.e. status epilepticus (SE), and repeated, brief seizures affect neuronal structure and function in both the immature and mature brain has been the subject of increasing clinical and experimental research. Earlier studies suggest that seizure-induced effects in the immature brain compared with the adult brain are different. This is manifested as differences in neuronal vulnerability, cellular and synaptic reorganization and regenerative processes. The focus of this review is first to give a short overview of currently used experimental models of epilepsy in immature rats, and then discuss more thoroughly seizure-induced acute and sub-acute cellular and molecular alterations, highlight the contribution of inflammatory-like reactions and intracellular cytoskeleton to the insult, and reveal changes in the structure and function of inhibitory GABA(A) and excitatory glutamate receptors. The role of seizure-activated reparative, plastic processes, synaptic remodelling, neurogenesis as well as the long-term consequences of seizures are briefly outlined. The main emphasis is put on studies carried out in experimental animals, and the focus of interest is the hippocampus, the brain area of great vulnerability in epilepsy. In vitro studies are discussed only to limited extent. Collectively, recent studies suggest that the deleterious effects of seizures may not solely be a consequence of neuronal damage and loss per se, but could be due to the fact that seizures interfere with the highly regulated developmental processes in the immature brain.


Subject(s)
Brain Damage, Chronic/physiopathology , Brain/growth & development , Brain/physiopathology , Epilepsy/physiopathology , Nerve Degeneration/physiopathology , Animals , Brain/metabolism , Brain Damage, Chronic/etiology , Brain Damage, Chronic/metabolism , Cell Differentiation/physiology , Disease Models, Animal , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/physiopathology , Epilepsy/metabolism , Humans , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Neuronal Plasticity/physiology , Rats , Receptors, Neurotransmitter/metabolism , Synapses/metabolism
14.
Brain Res ; 1187: 33-41, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18021757

ABSTRACT

Pre- and postnatal protein malnutrition (PMN) adversely affects the developing brain in numerous ways, but only a few studies have investigated specific glial parameters. This study aimed to evaluate specific glial changes of rats exposed to pre and postnatal PMN, based on glial fibrillary acidic protein (GFAP) and S100B immunocontents as well as glutamine synthetase (GS), in cerebral cortex, hippocampus, cerebellum and cerebrospinal fluid, on the 2nd, 15th and 60th postnatal days. We found increases in GFAP, S100B and GS in the cerebral cortex at birth, suggesting an astrogliosis. Hippocampus and cerebellum also exhibited this profile at birth. However, a significant interaction between age and diet in postnatal life was observed only in the S100B of the cerebral cortex. No changes in the content of GFAP and S100B and GS activity were found on the 60th postnatal day in malnourished rats. In contrast, following an increase in the levels of S100B in the cerebrospinal fluid, during the early developmental stages, levels remained elevated on the 60th postnatal day. Our data support the concept of astrogliosis at birth, induced by PMN, and involve extracellular-regulated kinase activation. Specific alterations in cerebral cortex emphasize the regional vulnerability of the brain to malnutrition; some alterations were observed only at birth (e.g. GFAP); others were observed on the 2nd and 15th post-natal days (e.g. ERK phosphorylation). Taken together, transient and persistent alterations (e.g. elevated extracellular levels of S100B) suggest some brain damage or a risk of brain diseases in rats exposed to PMN.


Subject(s)
Brain Damage, Chronic/etiology , Brain Damage, Chronic/physiopathology , Fetal Nutrition Disorders/physiopathology , Gliosis/etiology , Gliosis/physiopathology , Protein Deficiency/physiopathology , Aging/metabolism , Animals , Animals, Newborn , Biomarkers/analysis , Biomarkers/cerebrospinal fluid , Brain/embryology , Brain/growth & development , Brain/physiopathology , Brain Damage, Chronic/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Gliosis/metabolism , Glutamate-Ammonia Ligase/cerebrospinal fluid , Male , Nerve Growth Factors/cerebrospinal fluid , Neuroglia/metabolism , Pregnancy , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , S100 Proteins/cerebrospinal fluid
15.
J Neurosci ; 26(30): 7885-97, 2006 Jul 26.
Article in English | MEDLINE | ID: mdl-16870734

ABSTRACT

Brain size is precisely regulated during development and involves coordination of neural progenitor cell proliferation, differentiation, and survival. The adapter protein ShcA transmits signals from receptor tyrosine kinases via MAPK (mitogen-activated protein kinase)/ERK (extracellular signal-regulated kinase) and PI3K (phosphatidylinositol 3-kinase)/Akt signaling pathways. In the CNS, ShcA expression is high during embryonic development but diminishes as cells differentiate and switches to ShcB/Sck/Sli and ShcC/N-Shc/Rai. To directly test ShcA function in brain development, we used Cre/lox technology to express a dominant-negative form of ShcA (ShcFFF) in nestin-expressing neural progenitors. ShcFFF-expressing mice display microencephaly with brain weights reduced to 50% of littermate controls throughout postnatal and adult life. The cerebrum appeared most severely affected, but the gross architecture of the brain is normal. Body weight was mildly affected with a delay in reaching mature weight. At a mechanistic level, the ShcFFF microencephaly phenotype appears to be primarily attributable to elevated apoptosis levels throughout the brain from embryonic day 10.5 (E10.5) to E12, which declined by E14.5. Apoptosis remained at normal basal levels throughout postnatal development. Proliferation indices were not significantly altered in the embryonic neuroepithelium or within the postnatal subventricular zone. In another approach with the same nestin-Cre transgene, conditional deletion of ShcA in mice with a homozygous floxed shc1 locus also showed a similar microencephaly phenotype. Together, these data suggest a critical role for ShcA in neural progenitor survival signaling and in regulating brain size.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Brain Damage, Chronic/metabolism , Brain/embryology , Brain/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Brain/pathology , Brain Damage, Chronic/pathology , Cell Differentiation , Cell Survival , Mice , Mice, Inbred C57BL , Neurons/pathology , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Stem Cells/pathology
16.
Neuroscience ; 144(3): 920-33, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17161913

ABSTRACT

Rats raised in an enriched environment (enriched rats) have been reported to show less motor dysfunction following brain lesions, but the neuronal correlates of this improvement have not been well clarified. The present study aimed to elucidate the effect of chemical brain lesions and environmental enrichment on motor function and lesion-induced neurogenesis. Three week-old, recently weaned rats were divided into two groups: one group was raised in an enriched environment and the other group was raised in a standard cage for 5 weeks. Striatal damage was induced at an age of 8 weeks by injection of the neuro-toxins 6-hydroxydopamine (6-OHDA) or quinolinic acid (QA) into the striatum, or by injection of 6-OHDA into the substantia nigra (SN), which depleted nigrostriatal dopaminergic innervation. Enriched rats showed better performance on beam walking compared with those raised in standard conditions, but both groups showed similar forelimb use asymmetry in a cylinder test. The number of bromodeoxyuridine-labeled proliferating cells in the subventricular zone was increased by a severe striatal lesion induced by QA injection 1 week after the lesion, but decreased by injection of 6-OHDA into the SN. Following induction of lesions by striatal injection of 6-OHDA or QA, the number of cells positive for doublecortin (DCX) was strongly increased in the striatum; however, there was no change in the number of DCX-positive cells following 6-OHDA injection into the SN. Environmental enrichment enhanced the increase of DCX-positive cells with migrating morphology in the dorsal striatum. In enriched rats, DCX-positive cells traversed the striatal parenchyma far from the corpus callosum and lateral ventricle. DCX-positive cells co-expressed an immature neuronal marker, polysialylated neural cell adhesion molecule, but were negative for a glial marker. These data suggest that environmental enrichment improves motor performance on beam walking and enhances neuronal migration toward a lesion area in the striatum.


Subject(s)
Brain Damage, Chronic/therapy , Corpus Striatum/metabolism , Environment , Microtubule-Associated Proteins/metabolism , Nerve Regeneration/physiology , Neuropeptides/metabolism , Recovery of Function/physiology , Animals , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation , Corpus Striatum/cytology , Corpus Striatum/drug effects , Doublecortin Domain Proteins , Doublecortin Protein , Locomotion/physiology , Male , Motor Skills/physiology , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/physiology , Neurotoxins/pharmacology , Oxidopamine/pharmacology , Physical Stimulation , Quinolinic Acid/pharmacology , Rats , Rats, Wistar , Sialic Acids/metabolism , Stem Cells/cytology , Stem Cells/physiology
17.
Neurochem Int ; 51(6-7): 370-6, 2007.
Article in English | MEDLINE | ID: mdl-17513015

ABSTRACT

Mild to severe cognitive impairments are frequently observed symptoms in chronic alcoholics. Decline of cognitive function significantly affects patients' recovery process and prognosis. The hippocampal region is sensitive to the effects of alcohol and it has been suggested that alcohol-induced hippocampal damage and/or changes in neuronal circuitry play an important role in generating these symptoms. Although various hypotheses have been proposed, molecular mechanisms underlying these alterations in the hippocampus are largely unknown. In the present study, we employed a 2DE-based proteomics approach to compare the protein expression profiles of the hippocampus in human alcoholic and healthy control brains. In the alcoholic hippocampus, 20 protein spots were found to be differentially regulated, 2 increased and 18 decreased. Seventeen proteins were identified using mass spectroscopy and were subcategorized into three energy metabolic proteins, six protein metabolic proteins, four signalling proteins, two oxidative stress-related proteins, one vesicle trafficking protein and one cytoskeletal protein. Some of these proteins have been previously implicated in alcohol-induced brain pathology. Based upon the results, several hypotheses were generated to explain the mechanisms underlying possible functional and/or structural alterations induced by chronic alcohol use in this brain region.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Alcoholism/complications , Brain Damage, Chronic/chemically induced , Ethanol/adverse effects , Hippocampus/drug effects , Nerve Tissue Proteins/drug effects , Adult , Aged , Aged, 80 and over , Alcohol-Induced Disorders, Nervous System/physiopathology , Alcoholism/physiopathology , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Central Nervous System Depressants/adverse effects , Electrophoresis, Gel, Two-Dimensional , Energy Metabolism/drug effects , Energy Metabolism/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Male , Middle Aged , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protein Transport/drug effects , Protein Transport/physiology , Proteomics , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Neurosci Res ; 59(3): 277-87, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17765347

ABSTRACT

The tissue distribution of glial cell line-derived neurotrophic factor (GDNF) during development and changes in GDNF levels by unilateral 6-hydroxydopamine lesions were investigated in rats using a newly established enzyme immunoassay system and by immunohistochemistry. The detection limit of the assay was 0.3 pg/0.2 ml and the system recognized glycosylated mature GDNF. Concentrations of GDNF were relatively high in the kidney and testis during the embryonic and neonatal periods, respectively, and decreased with age. In the striatum, hippocampus and brain stem, GDNF reached a maximal level at around postnatal day 14. However, brain levels were generally lower than those in non-neural tissues. In the CNS, GDNF immunoreactivity was observed in striatal neurons, pyramidal neurons in the hippocampus and the Vth layer of the cortex, large neurons in the diagonal band and brain stem, and spinal motor neurons. It was also evident in several non-neural, tissue-specific cells, such as cells in the renal collecting ducts and distal tubules, and testicular Sertoli cells. Destruction of nigral dopaminergic neurons by 6-hydroxydopamine enhanced the levels of striatal GDNF protein, with apparent involvement of astrocytes. These results suggest that GDNF is normally synthesized in neurons, but may also be produced by astroglial cells in damaged brains.


Subject(s)
Astrocytes/metabolism , Brain/embryology , Brain/growth & development , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Neurons/metabolism , Animals , Animals, Newborn , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Brain Mapping , Denervation , Glial Cell Line-Derived Neurotrophic Factor/analysis , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Immunoassay , Immunohistochemistry , Kidney/enzymology , Kidney/growth & development , Male , Nerve Regeneration/physiology , Neurotoxins , Oxidopamine , Rats , Rats, Sprague-Dawley , Testis/embryology , Testis/growth & development , Up-Regulation/physiology
19.
Brain Res ; 1131(1): 77-87, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17184742

ABSTRACT

Insulin-like growth factor-2 (IGF2) is a member of the insulin gene family with known neurotrophic properties. The actions of IGF2 are mediated via the IGF type 1 and type 2 receptors as well as through the insulin receptors, all of which are widely expressed throughout the brain. Since IGF2 is up-regulated in the brain after injury, we wanted to determine whether the absence of IGF2 can lead to any alteration on brain morphology and/or in the response of its receptor binding sites following a neurotoxic insult. No morphological differences were observed between the brains of IGF2 knockout (IGF2(-/-)) and wild-type control (IGF2(+/+)) mice. However, our in vitro receptor autoradiography results indicate that IGF2(-/-) mice had lower endogenous levels of [(125)I]IGF1 and [(125)I]insulin receptor binding sites in the hippocampus and cerebellum as compared to IGF2(+/+) mice, while endogenous [(125)I]IGF2 receptor binding showed a decrease only in the cerebellum. Seven days after kainic acid administration, the [(125)I]insulin receptor binding sites were significantly decreased in all brain regions of the IGF2(+/+) mice, while the levels of [(125)I]IGF1 and [(125)I]IGF2 binding sites were decreased only in select brain areas. The IGF2(-/-) mice, on the other hand, showed increased [(125)I]IGF1 and [(125)I]IGF2 and [(125)I]insulin receptor binding sites in selected regions such as the hippocampus and cerebellum. These results, taken together, suggest that deletion of IGF2 gene does not affect gross morphology of the brain but does selectively alter endogenous [(125)I]IGF1, [(125)I]IGF2 and [(125)I]insulin receptor binding sites and their response to neurotoxicity.


Subject(s)
Binding, Competitive/drug effects , Brain/metabolism , Insulin-Like Growth Factor II/genetics , Neurotoxins/pharmacology , Receptor, IGF Type 2/metabolism , Age Factors , Animals , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/physiology , Brain/drug effects , Brain/embryology , Brain Damage, Chronic/genetics , Brain Damage, Chronic/metabolism , Brain Damage, Chronic/physiopathology , Cerebellum/drug effects , Cerebellum/embryology , Cerebellum/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Resistance/genetics , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Iodine Radioisotopes , Kainic Acid/pharmacology , Male , Mice , Mice, Knockout , Radioligand Assay , Receptor, IGF Type 2/drug effects , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism
20.
Brain Res ; 1133(1): 209-15, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17196560

ABSTRACT

Cardiac arrest-induced cerebral hypoxic injury could induce posthypoxic movement disorders. Here we investigated the effects of memantine, an NMDA receptor channel blocker, on the neurodegeneration occurred in an established rat model of posthypoxic myoclonus. We found that administration of memantine for 7 days significantly reduced cerebral hypoxia-induced neurodegeneration in the CA1 of the hippocampus, the reticular thalamic nucleus (RTN) and the primary fissure of the cerebellum of the posthypoxic animals. The results suggest that the neurodegeneration observed in specific areas of the brain of the posthypoxic rats is contributed by NMDA receptor-mediated excitotoxicity.


Subject(s)
Brain Damage, Chronic/metabolism , Hypoxia, Brain/metabolism , Myoclonus/metabolism , Nerve Degeneration/metabolism , Neurotoxins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/physiopathology , Cerebellum/drug effects , Cerebellum/metabolism , Cerebellum/physiopathology , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Hypoxia, Brain/complications , Hypoxia, Brain/physiopathology , Intralaminar Thalamic Nuclei/drug effects , Intralaminar Thalamic Nuclei/metabolism , Intralaminar Thalamic Nuclei/physiopathology , Male , Memantine/pharmacology , Myoclonus/etiology , Myoclonus/physiopathology , Nerve Degeneration/physiopathology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL