Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Mol Ther ; 25(5): 1155-1162, 2017 05 03.
Article En | MEDLINE | ID: mdl-28389320

Lysosomal storage disorders (LSDs) are a broad class of monogenic diseases with an overall incidence of 1:7,000 newborns, due to the defective activity of one or more lysosomal hydrolases or related proteins resulting in storage of un-degraded substrates in the lysosomes. The over 40 different known LSDs share a life-threatening nature, but they are present with extremely variable clinical manifestations, determined by the characteristics and tissue distribution of the material accumulating due to the lysosomal dysfunction. The majority of LSDs lack a curative treatment. This is particularly true for LSDs severely affecting the CNS. Based on current preclinical and clinical evidences, among other treatment modalities, hematopoietic stem cell gene therapy could potentially result in robust therapeutic benefit for LSD patients, with particular indication for those characterized by severe brain damage. Optimization of current approaches and technology, as well as implementation of clinical trials for novel indications, and prolonged and more extensive follow-up of the already treated patients will allow translating this promise into new medicinal products.


Brain Damage, Chronic/therapy , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hydrolases/genetics , Lysosomal Storage Diseases/therapy , Animals , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/genetics , Brain Damage, Chronic/pathology , Disease Models, Animal , Enzyme Replacement Therapy/methods , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hematopoietic Stem Cells/cytology , Humans , Hydrolases/deficiency , Lentivirus/genetics , Lentivirus/metabolism , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Lysosomes/metabolism , Transplantation, Homologous
2.
Mol Neurobiol ; 52(3): 1527-1539, 2015 Dec.
Article En | MEDLINE | ID: mdl-25367879

This study aims to clarify the potential role of Raf-1 kinase in cerebral vasospasm (CVS) and early brain injury (EBI) after subarachnoid hemorrhage (SAH). Two experimental SAH models in rats, including cisterna magna double injection model for CVS study and prechiasmatic cistern single injection model for EBI study, were performed in this research. As a specific inhibitor of Raf-1, BAY 43-9006 was used in this study. In CVS study, time course study showed that the basilar artery exhibited vasospasm after SAH and became most severe at day 5, and the phosphorylation of Raf-1 had the same trends, while both vasospasm and the phosphorylation of Raf-1 induced by SAH were inhibited by BAY 43-9006 treatment. In addition, BAY 43-9006 treatment significantly reversed the phosphorylation of ERK1/2 and the activation of NF-κB induced by SAH and decreased the messenger RNA (mRNA) levels of IL-6 and IL-1ß. In EBI study, BAY 43-9006 treatment significantly suppressed the brain injury induced by SAH. Besides, BAY 43-9006 inhibited the phosphorylation of Raf-1 and ERK1/2; decreased the protein levels of COX-2, VEGF, and MMP-9; and reversed the activation of NF-κB induced by SAH. These results demonstrate that Raf-1 kinase contributes to CVS and EBI after SAH by enhancing the activation of the Raf-1/ERK1/2 and Raf-1/NF-κB signaling pathways, and that the inhibition of these pathways might offer new treatment strategies for CVS and EBI.


Brain Damage, Chronic/enzymology , MAP Kinase Kinase Kinases/physiology , Nerve Tissue Proteins/physiology , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/enzymology , Animals , Basilar Artery/physiopathology , Basilar Artery/ultrastructure , Blood-Brain Barrier , Brain Damage, Chronic/etiology , Brain Edema/etiology , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Disease Models, Animal , Down-Regulation/drug effects , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Mitogen-Activated Protein Kinase Kinases/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-raf , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sorafenib , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vasospasm, Intracranial/etiology
3.
Brain Res ; 1587: 15-22, 2014 Oct 31.
Article En | MEDLINE | ID: mdl-25199591

Early brain injury (EBI) plays a key role in the pathogenesis of subarachnoid hemorrhage (SAH). Although the neuroprotective effects of ghrelin have been demonstrated in several studies, whether ghrelin reduces EBI after SAH remains unknown. In this study, we hypothesized that treatment with ghrelin would attenuate EBI after SAH, and that this protection would be mediated, at least in part, by activation of the PI3K/Akt signaling pathway. Adult male Sprague-Dawley rats (n=100) were randomly divided into the following groups: control group (n=20), SAH group (n=20), SAH+vehicle group (n=20), SAH+ghrelin group (n=20) and SAH+ghrelin+LY294002 group (n=20). The rats were injected with autologous blood (0.3mL) into the prechiasmatic cistern to induce SAH. Ghrelin (80µg/kg, IP), or an equal volume of vehicle, was administered immediately after surgery. The PI3K inhibitor, LY294002, was applied to manipulate the proposed pathway. Mortality, neurological scores, brain edema, cell apoptosis, and the expression of p-Akt, and cleaved caspase-3 proteins were assayed after 24h SAH. Ghrelin significantly improved neurological function and reduced neuronal apoptosis and brain edema at 24h after SAH. The level of p-Akt, expressed mainly in neurons, was markedly up-regulated. Additionally, the level of cleaved caspase-3 was decreased by ghrelin treatment. The beneficial effects of ghrelin in SAH rats were partially suppressed by LY294002. These results demonstrate that ghrelin may reduce EBI after SAH, via a mechanism involving the PI3K/Akt signaling pathway.


Brain Damage, Chronic/prevention & control , Brain Edema/prevention & control , Ghrelin/therapeutic use , Nerve Tissue Proteins/physiology , Neuroprotective Agents/therapeutic use , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Subarachnoid Hemorrhage/drug therapy , Animals , Apoptosis/drug effects , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/pathology , Brain Edema/enzymology , Brain Edema/etiology , Caspase 3/metabolism , Chromones/pharmacology , Enzyme Induction/drug effects , Ghrelin/pharmacology , Male , Morpholines/pharmacology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurologic Examination , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Single-Blind Method , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology , Up-Regulation/drug effects
4.
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
5.
Brain Res ; 1398: 86-93, 2011 Jun 29.
Article En | MEDLINE | ID: mdl-21172324

The present study investigates the role of protein kinase C alpha/delta (PKCα/PKCδ) in brain injury induced by intracerebral hemorrhage (ICH) by utilizing a rat model that received intracerebral injections of autologous blood and thrombin (TM). The activation and expression of PKC and PKCδ were analyzed by Western blot and immunohistochemistry. A PKC inhibitor, dihydrochloride (H7), was administrated intraperitoneally after injury to evaluate the effect of inhibition of PKC on ICH and TM induced brain damage. Our data indicate that both ICH and TM increased the expression of PKCα/PKCδ in the brain tissue, and PKCα expression peaked at 6h, while PKCδ expression reached its maximum value at 72h post-injury. Administration of H7 significantly reduced the inflammatory cells infiltrate, permeability of brain-blood barrier (BBB), brain edema, and neuronal death. We conclude that both PKCα and PKCδ play important roles in ICH and TM-induced brain injury, and dihydrochloride (H7) can attenuate brain damage after ICH.


Brain Damage, Chronic/enzymology , Brain Damage, Chronic/physiopathology , Cerebral Hemorrhage/enzymology , Cerebral Hemorrhage/physiopathology , Protein Kinase C-alpha/physiology , Protein Kinase C-delta/physiology , Signal Transduction/physiology , Thrombin/toxicity , Animals , Brain Damage, Chronic/pathology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Male , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-delta/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Thrombin/metabolism
6.
Int J Biochem Cell Biol ; 41(4): 717-20, 2009 Apr.
Article En | MEDLINE | ID: mdl-18490188

Thiamin, or vitamin B1, is crucial for brain function. In its active form, thiamin pyrophosphate (TPP), it is a co-enzyme for several enzymes, including transketolase. Transketolase is an important enzyme in the non-oxidative branch of the pentose phosphate pathway (PPP), a pathway responsible for generating reducing equivalents, which is essential for energy transduction and for generating ribose for nucleic acid synthesis. Transketolase also links the PPP to glycolysis, allowing a cell to adapt to a variety of energy needs, depending on its environment. Abnormal transketolase expression and/or activity have been implicated in a number of diseases where thiamin availability is low, including Wernicke-Korsakoff's Syndrome and alcoholism. Yet, the precise mechanism by which this enzyme is involved in the pathophysiology of these disorders remains controversial.


Alcoholism/enzymology , Brain Damage, Chronic/enzymology , Transketolase/metabolism , Brain Damage, Chronic/chemically induced , Humans , Korsakoff Syndrome/enzymology , Thiamine Deficiency/enzymology , Thiamine Pyrophosphate/deficiency
7.
Exp Neurol ; 213(1): 145-53, 2008 Sep.
Article En | MEDLINE | ID: mdl-18599042

Brain cell vulnerability to neurologic insults varies greatly, depending on their neuronal subpopulation. Among cells that survive a pathological insult such as ischemia or brain trauma, some may undergo morphological and/or biochemical changes that could compromise brain function. We previously reported that surviving cortical GABAergic neurons exposed to glutamate in vitro displayed an NMDA receptor (NMDAR)-mediated alteration in the levels of the GABA synthesizing enzyme glutamic acid decarboxylase (GAD65/67) [Monnerie, H., Le Roux, P., 2007. Reduced dendrite growth and altered glutamic acid decarboxylase (GAD) 65- and 67-kDa isoform protein expression from mouse cortical GABAergic neurons following excitotoxic injury in vitro. Exp. Neurol. 205, 367-382]. In this study, we examined the mechanisms by which glutamate excitotoxicity caused a change in cortical GABAergic neurons' GAD protein levels. Removing extracellular calcium prevented the NMDAR-mediated decrease in GAD protein levels, measured using Western blot techniques, whereas inhibiting calcium entry through voltage-gated calcium channels had no effect. Glutamate's effect on GAD protein isoforms was significantly attenuated by preincubation with the cysteine protease inhibitor N-Acetyl-L-Leucyl-L-Leucyl-L-norleucinal (ALLN). Using class-specific protease inhibitors, we observed that ALLN's effect resulted from the blockade of calpain and cathepsin protease activities. Cell-free proteolysis assay confirmed that both proteases were involved in glutamate-induced alteration in GAD protein levels. Together these results suggest that glutamate-induced excitotoxic stimulation of NMDAR in cultured cortical neurons leads to altered GAD protein levels from GABAergic neurons through intracellular calcium increase and protease activation including calpain and cathepsin. Biochemical alterations in surviving cortical GABAergic neurons in various disease states may contribute to the altered balance between excitation and inhibition that is often observed after injury.


Cerebral Cortex/enzymology , Cysteine Endopeptidases/metabolism , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Neurons/enzymology , gamma-Aminobutyric Acid/metabolism , Animals , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/physiopathology , Calcium Signaling/drug effects , Calpain/metabolism , Cathepsins/metabolism , Cells, Cultured , Cerebral Cortex/physiopathology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation/drug effects , Epilepsy/enzymology , Epilepsy/physiopathology , Glutamate Decarboxylase/drug effects , Glutamic Acid/toxicity , Hypoxia-Ischemia, Brain/enzymology , Hypoxia-Ischemia, Brain/physiopathology , Mice , Mice, Inbred BALB C , Neurons/drug effects , Neurotoxins/metabolism , Neurotoxins/toxicity , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Brain Res ; 1221: 134-40, 2008 Jul 24.
Article En | MEDLINE | ID: mdl-18554575

Glutamate is the main excitatory neurotransmitter in brain involved in pathophysiology of several brain injuries. In this context, glutamate showed to stimulate ecto-nucleotidase activities in cerebellar granule cells increasing extracellular adenosine levels, an important neuromodulator in the CNS able to prevent cell damage. The organoselenium compounds, such as ebselen and diphenyl diselenide [(PhSe)(2)], display neuroprotective activities mediated at least in part by their antioxidant and anti-inflammatory properties. Ebselen was described to prevent glutamate-induced lipid peroxidation and cell death in cerebellar granule cells and (PhSe)(2) modify glutamatergic synapse parameters in vitro and in vivo. In the present study, we investigated the effects of ebselen or (PhSe)(2) on glutamate-induced stimulation of ecto-nucleotidase activities in rat cultured cerebellar granule cells. Glutamate increased nucleotide hydrolysis at lower concentrations (10 and 100 microM) than described in the literature and this effect was counteracted by both organoselenium compounds tested. Based on these results, we investigated the association of organoselenium effects with their antioxidant properties searching for redox site modulation by using the alkylant agent N-ethylmaleimide (NEM). Our results suggest that selenium compounds, as well as the well-known antioxidant trolox, can avoid the increase on glutamate-induced stimulation of ecto-nucleotidase activities probably due to their antioxidant properties.


Adenosine Triphosphatases/drug effects , Brain Damage, Chronic/drug therapy , Nerve Degeneration/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Selenium/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Antioxidants/pharmacology , Azoles/pharmacology , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/physiopathology , Cells, Cultured , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/metabolism , Chromans/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , Glutamic Acid/metabolism , Isoindoles , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Neurons/enzymology , Neuroprotective Agents/metabolism , Nucleotides/metabolism , Organoselenium Compounds/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Selenium/metabolism
9.
J Child Neurol ; 22(10): 1214-21, 2007 Oct.
Article En | MEDLINE | ID: mdl-17940249

Isolated sulfite oxidase deficiency is a rare genetic neurometabolic disease. The first symptoms of this disorder (similar to symptoms of ischemic events) may lead to misdiagnosis and to subsequent birth of affected children in these families. This study characterizes the magnetic resonance (MR) imaging and (for the first time, to our knowledge) the MR spectroscopy features of isolated sulfite oxidase deficiency to provide a means for early and correct diagnosis. Three patients with isolated sulfite oxidase deficiency are studied who manifested intractable seizures and severe hypotonia in the immediate postnatal period with an unknown diagnosis, despite extensive workup. MR imaging and proton MR spectroscopy examinations were performed early in the neonatal period in 2 infants and after 5 months in the third infant. The prominent MR features were early cystic white matter damage, accompanied by profound cerebral atrophy in the third infant. Compared with hypoxic-ischemic disorder, MR findings in isolated sulfite oxidase deficiency demonstrate a more severe condition, without subsequent recovery. The MR spectroscopy studies indicate early onset of energetic and metabolic imbalance. Urine stick findings demonstrated high sulfite levels in 2 patients, and the final diagnosis was subsequently made based on molecular, biochemical, and genetic findings. Magnetic resonance imaging and MR spectroscopy measurements may help differentiate isolated sulfite oxidase deficiency from hypoxic-ischemic condition in patients in whom this diagnosis is not clinically suspected and may lead to further genetic antenatal inquiry that might prevent the birth of other infants affected with this severe and incurable congenital disease.


Brain Diseases, Metabolic, Inborn/pathology , Brain/pathology , Genetic Predisposition to Disease/genetics , Oxidoreductases Acting on Sulfur Group Donors/deficiency , Oxidoreductases Acting on Sulfur Group Donors/genetics , Amino Acids, Sulfur/metabolism , Atrophy/enzymology , Atrophy/genetics , Atrophy/physiopathology , Brain/enzymology , Brain/physiopathology , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/genetics , Brain Damage, Chronic/physiopathology , Brain Diseases, Metabolic, Inborn/enzymology , Brain Diseases, Metabolic, Inborn/physiopathology , Diagnosis, Differential , Early Diagnosis , Epilepsy/enzymology , Epilepsy/genetics , Epilepsy/physiopathology , Female , Genetic Markers/genetics , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Sulfites/urine
10.
Can J Neurol Sci ; 34(3): 356-61, 2007 Aug.
Article En | MEDLINE | ID: mdl-17803037

BACKGROUND: Oxidative stress largely contributes to early brain injury after subarachnoid hemorrhage (SAH). One of the major sources of reactive oxygen species is NADPH oxidase, upregulated after SAH. We hypothesized that NADPH oxidase-induced oxidative stress plays a major causative role in early brain injury after SAH. METHODS: Using gp91phox knockout (ko) and wild-type (wt) mice, we studied early brain injury in the endovascular perforation model of SAH. Mortality rate, cerebral edema, oxidative stress, and superoxide production were measured at 24 h after SAH. Neurological evaluation was done at 23 h after SAH surgery. RESULTS: Genotyping confirmed the existence of a nonfunctional gp91phox gene in the ko mice. CBF measurements did not show differences in SAH-induced acute ischemia between ko and wt mice. SAH caused a significant increase of water content in the ipsilateral hemisphere as well as an increase of Malondialdehyde (MDA) levels and superoxide production. There were no significant differences in post-SAH mortality rate, brain water content and the intensity of the oxidative stress between knockout and wild type groups of mice. CONCLUSIONS: Our results suggest that gp91phox is not critically important to the early brain injury after SAH. An adaptive compensatory mechanism for free radical production in knockout mice is discussed.


Brain Damage, Chronic/genetics , Membrane Glycoproteins/genetics , NADPH Oxidases/genetics , Nerve Degeneration/genetics , Oxidative Stress/genetics , Subarachnoid Hemorrhage/complications , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/physiopathology , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/physiopathology , Brain Edema/enzymology , Brain Edema/genetics , Brain Edema/physiopathology , Cerebral Arteries/enzymology , Cerebral Arteries/physiopathology , Free Radicals/metabolism , Genetic Markers/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/physiopathology , Superoxides/metabolism
11.
Glia ; 55(9): 954-65, 2007 Jul.
Article En | MEDLINE | ID: mdl-17487878

Caspase-3 has classically been defined as the main executioner of programmed cell death. However, recent data supports the participation of this protease in non-apoptotic cellular events including cell proliferation, cell cycle regulation, and cellular differentiation. In this study, astroglial cleavage of caspase-3 was analyzed following excitotoxic damage in postnatal rats to determine if its presence is associated with apoptotic cell death, cell proliferation, or cytoskeletal remodeling. A well-characterized in vivo model of excitotoxicity was studied, where damage was induced by intracortical injection of N-methyl-D-asparate (NMDA) in postnatal day 9 rats. Our results demonstrate that cleaved caspase-3 was mainly observed in the nucleus of activated astrocytes in the lesioned hemisphere as early as 4 h postlesion and persisted until the glial scar was formed at 7-14 days, and it was not associated with TUNEL labeling. Caspase-3 enzymatic activity was detected at 10 h and 1 day postlesion in astrocytes, and co-localized with caspase-cleaved fragments of glial fibrillary acidic protein (CCP-GFAP). However, at longer survival times, when astroglial hypertrophy was observed, astroglial caspase-3 did not generally correlate with GFAP cleavage, but instead was associated with de novo expression of vimentin. Moreover, astroglial caspase-3 cleavage was not associated with BrdU incorporation. These results provide further evidence for a nontraditional role of caspases in cellular function that is independent of cell death and suggest that caspase activation is important for astroglial cytoskeleton remodeling following cellular injury.


Astrocytes/enzymology , Brain Damage, Chronic/enzymology , Brain/enzymology , Caspase 3/metabolism , Cytoskeleton/enzymology , Gliosis/chemically induced , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/pathology , Brain/pathology , Brain/physiopathology , Brain Damage, Chronic/physiopathology , Cell Death/drug effects , Cell Death/physiology , Cell Proliferation/drug effects , Cell Size/drug effects , Cytoskeleton/drug effects , Cytoskeleton/pathology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/enzymology , Gliosis/physiopathology , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Male , N-Methylaspartate/metabolism , N-Methylaspartate/toxicity , Nerve Degeneration/chemically induced , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Neurotoxins/metabolism , Neurotoxins/toxicity , Rats , Rats, Long-Evans , Vimentin/metabolism
12.
Stroke ; 37(11): 2816-23, 2006 Nov.
Article En | MEDLINE | ID: mdl-17008622

BACKGROUND AND PURPOSE: Cerebrovascular white matter (WM) lesions contribute to cognitive impairment and motor dysfunction in the elderly. A disruption of the blood-brain barrier (BBB) is believed to be a critical early event leading to these WM lesions. Previous studies have suggested the involvement of matrix metalloproteinase-2 (MMP-2) in BBB disruptions and the upregulation of MMP-2 after chronic cerebral hypoperfusion in a rat model. In the present study, we asked whether MMP-2 is involved in the BBB disruption and the subsequent WM lesions after chronic cerebral hypoperfusion. METHODS: We compared the severity of white matter lesions in rats after chronic cerebral hypoperfusion with or without an MMP inhibitor. Then, we also induced the chronic cerebral hypoperfusion in wild-type and MMP-2-null mice. RESULTS: In the rats treated with a relatively selective MMP-2 inhibitor, AG3340, the WM lesions after chronic cerebral hypoperfusion were significantly less severe, and the number of activated astroglia and microglia were also significantly lower as compared with the vehicle-treated rats. Gene knockout of MMP-2 also reduced the severity of the WM lesions and the number of activated astroglia and microglia in a mice system. In both rodents, the disruption of BBB function, as assessed by IgM staining and the Evans blue extravasation test, was less severe when MMP-2 activity was attenuated. CONCLUSIONS: These findings indicate that MMP-2 plays a critical role in the BBB disruption, glial cell activation, and WM lesions after chronic cerebral hypoperfusion and suggest the potential value of MMP-2 inhibitors as a therapeutic tool in cerebrovascular WM lesions.


Brain Damage, Chronic/enzymology , Brain/blood supply , Brain/enzymology , Matrix Metalloproteinase 2/physiology , Nerve Fibers, Myelinated/enzymology , Animals , Brain/pathology , Brain Damage, Chronic/pathology , Male , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Fibers, Myelinated/pathology , Rats , Rats, Wistar , Severity of Illness Index
13.
Brain Res ; 1064(1-2): 108-18, 2005 Dec 07.
Article En | MEDLINE | ID: mdl-16289484

Mild hypothermia improves survival and neurological outcome after cardiac arrest, as well as increasing activation of the extracellular-signal-regulated kinase (ERK) in hippocampus. ERK signaling is involved in neuronal growth and survival. We tested the hypothesis that the beneficial effects of hypothermia required ERK activation. ERK activation was measured by immunoblotting with phosphorylation-specific antibodies. Rats (n = 8 per group) underwent 8 min of asphyxial cardiac arrest and were resuscitated with chest compressions, ventilation, epinephrine and bicarbonate. At 30 min after resuscitation, vehicle (50% saline:50% DMSO) or the ERK kinase inhibitor U0126 (100 microg) was infused into the lateral ventricle. Cranial temperature was kept at either 33 degrees C (hypothermia) or 37 degrees C (normothermia) between 1 and 24 h. Neurological function was assessed daily for 14 days. Surviving neurons were counted in the hippocampus. A dose of 100 mug U0126 inhibited ERK bilaterally for 12 to 24 h and decreased phosphorylation of the ERK substrates ATF-2 and CREB. As in previous studies, hypothermia improved survival, neurological and histological outcome after cardiac arrest. However, survival, neurological score and histology did not differ between U0126 and vehicle-treated rats after cardiac arrest. Therefore, a dose of U0126 sufficient to inhibit biochemical markers of ERK signaling in hippocampus does not alter the beneficial effects of hypothermia induced after resuscitation in rats and did not affect recovery of normothermia-treated rats. These results suggest that hypothermia-induced improvement in outcomes does not require ERK activation.


Extracellular Signal-Regulated MAP Kinases/metabolism , Heart Arrest/enzymology , Heart Arrest/therapy , Hippocampus/enzymology , Hypothermia, Induced , Hypothermia/enzymology , Animals , Asphyxia/complications , Asphyxia/enzymology , Asphyxia/therapy , Body Temperature , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/etiology , Brain Damage, Chronic/prevention & control , Butadienes/administration & dosage , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Heart Arrest/complications , Hypothermia/complications , Injections, Intraventricular , Male , Nitriles/administration & dosage , Rats , Rats, Sprague-Dawley , Resuscitation , Signal Transduction/physiology
14.
Brain Res Mol Brain Res ; 137(1-2): 202-12, 2005 Jun 13.
Article En | MEDLINE | ID: mdl-15950779

Glial activation and inflammation following brain injury may initiate and maintain the process of neurodegeneration. Both glia and neurons synthesize proinflammatory mediators such as interleukin 1 beta (IL-1beta), cytosolic phospholipase A2 (cPLA2), cyclooxygenase-2 (COX-2), and prostaglandins. The molecular mechanisms by which IL-1beta regulates inflammatory genes such as cPLA2 and COX-2 in glial and neuronal cells are poorly understood. We have studied IL-1beta-mediated gene regulation in an established glial and neuronal human cell lines. We report that IL-1beta induced cPLA2 and COX-2 mRNA and protein expression and subsequent prostaglandin E2 (PGE2) release in a time-dependent manner in H4 neuroglioma cells. Both SB203580 and PD98059 [p38 and p42/44 mitogen-activated protein kinase (MAPKs) inhibitors, respectively] reduced IL-1beta-induced PGE2 production, while only SB203580 reduced both cPLA2 and COX-2 expression. Similarly, in SKNSH neuroblastoma cells, both SB203580 and PD98059 reduced IL-1beta-induced PGE2 release, with no detectable COX-2 and cPLA2 protein expression in these cells. Our results indicate that the signaling mechanisms of p38 and p42/44 MAPKs play a role in IL-1beta-mediated PGE2 release in both of these cell lines, with differences upstream at the level of cPLA(2)/COX-2 expression. IL-1beta-induced cPLA2 and COX-2 gene expression is modulated through the p38 MAPK pathway in both neuroglioma and neuroblastoma cells. Understanding the signaling mechanisms involved in IL-1beta-mediated inflammatory processes in both glia and neuronal cells may provide potential targets for therapeutic intervention for neurological disorders.


Brain/enzymology , Encephalitis/enzymology , Interleukin-1/metabolism , MAP Kinase Signaling System/physiology , Neurons/enzymology , Prostaglandin-Endoperoxide Synthases/genetics , Brain/immunology , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/physiopathology , Cell Line, Tumor , Cyclooxygenase 2 , Dinoprostone/metabolism , Encephalitis/physiopathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Glioma , Group IV Phospholipases A2 , Humans , Interleukin-1/pharmacology , MAP Kinase Signaling System/drug effects , Membrane Proteins , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Neuroblastoma , Neurons/drug effects , Neurons/immunology , Phospholipases A/genetics , Phospholipases A2 , RNA, Messenger/drug effects , RNA, Messenger/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Exp Neurol ; 185(1): 109-19, 2004 Jan.
Article En | MEDLINE | ID: mdl-14697322

In the developing central nervous system, a src-related protein-tyrosine kinase fyn participates in the myelination process, neuronal growth, and cytoskeletal organization. In adults, fyn has been implicated in learning and memory formation. To test if fyn expression is modulated by neuronal activity, we performed quantitative in situ hybridization (ISH) using brain sections of the adult rats that had undergone either kainic acid (KA)-induced seizures or neuronal deafferentation (entorhinal cortex lesion, ECL). In the KA model, a few hours after seizure activities, fyn mRNA was elevated in the dentate gyrus (DG) (+45%), cerebral cortex layer III (+35%), and piriform cortex (+25%). Conversely, fyn mRNA consistently decreased in the hippocampal neurons after transection of the major axonal inputs from the entorhinal cortex. Although fyn expression in the brain has been allegedly limited to neurons and oligodendrocytes, we provide in this study the first evidence that fyn mRNA is highly expressed in the astrocytes involved in reactive gliosis. In the KA model, the occurrence of fyn-overexpressing astrocytes increased with the progress of neuronal damage in the CA1 and CA3 regions of the hippocampus. In contrast, fyn-overexpressing astrocytes were not observed in the granular cell layer of dentate gyrus (DG), where neurons were not damaged. Likewise, in the ECL model, the most drastic change in fyn mRNA expression took place at the reactive astrocytes near the stab wound sites, where fyn mRNA levels were doubled 4-10 d after the lesion. Collectively, our data suggest that (i) an early induction of fyn mRNA in neurons is linked to neuronal activity, and (ii) the delayed induction of fyn mRNA in reactive astrocytes near the damaged cells may play novel signaling roles during glial response.


Astrocytes/enzymology , Brain Damage, Chronic/enzymology , Neurons/enzymology , Proto-Oncogene Proteins/metabolism , Seizures/enzymology , Animals , Astrocytes/pathology , Blotting, Western , Brain Damage, Chronic/pathology , Brain Damage, Chronic/physiopathology , Cell Death , Cells, Cultured , Disease Models, Animal , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Enzyme Induction , Kainic Acid , Male , Mice , Neurons/pathology , Organ Specificity , Perforant Pathway/physiopathology , Perforant Pathway/surgery , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fyn , RNA Probes/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/pathology
18.
Arterioscler Thromb Vasc Biol ; 23(2): 322-7, 2003 Feb 01.
Article En | MEDLINE | ID: mdl-12588778

OBJECTIVE: In the present study, MRI has been used to investigate therapeutic intervention with statins in a model of permanent focal cerebral ischemia in rat. METHODS AND RESULTS: Brain ischemia was induced in rats by the permanent occlusion of middle cerebral artery (MCAO) and the brain infarct size followed up in alive animals 2, 24, and 48 hours after MCAO, using the trace of apparent diffusion coefficient [Tr(D)] maps and T2-weighted images. In vehicle-treated rats, the infarct volumes increased by 38.5% and 89% after 24 and 48 hours, respectively, compared with the damage detected at 2 hours after MCAO. Treatment with simvastatin (20 mg/kg) after MCAO prevented the increase in brain infarct volume occurring at 24 hours and induced a 46.6% reduction after 48 hours. This effect was similar to that observed when simvastatin was administered before the induction of focal ischemia. T2W-MRI images confirmed these findings. The neuroprotective effects of simvastatin were paralleled by an increase in endothelial NO synthase immunoreactivity, detectable in the brain of simvastatin-treated rats. CONCLUSIONS: Statins, in addition to their preventive effect on cerebral ischemia, exert a neuroprotective role in the attenuation of brain damage after acute stroke.


Brain Damage, Chronic/prevention & control , Brain Injuries/drug therapy , Brain Ischemia/drug therapy , Simvastatin/therapeutic use , Animals , Brain Chemistry/drug effects , Brain Chemistry/physiology , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/pathology , Brain Injuries/complications , Brain Injuries/enzymology , Brain Injuries/pathology , Brain Ischemia/complications , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain Mapping/methods , Cerebral Arteries/drug effects , Cerebral Arteries/enzymology , Cerebral Arteries/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Cerebral Infarction/drug therapy , Cerebral Infarction/enzymology , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunohistochemistry/methods , Magnetic Resonance Imaging/methods , Male , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
19.
Curr Opin Infect Dis ; 15(3): 253-7, 2002 Jun.
Article En | MEDLINE | ID: mdl-12015459

In spite of improved antimicrobial therapy, bacterial meningitis still results in brain damage leading to significant long-term neurological sequelae in a substantial number of survivors, as confirmed by several recent studies. Meningitis caused by Streptococcus pneumoniae is associated with a particularly severe outcome. Experimental studies over the past few years have increased our understanding of the molecular mechanisms underlying the events that ultimately lead to brain damage during meningitis. Necrotic damage to the cerebral cortex is at least partly mediated by ischemia and oxygen radicals and therefore offers a promising target for adjunctive therapeutic intervention. Neuronal apoptosis in the hippocampus may represent the major pathological process responsible for cognitive impairment and learning disabilities in survivors. However, the mechanisms involved in causing this damage remain largely unknown. Anti-inflammatory treatment with corticosteroids aggravates hippocampal damage, thus underlining the potential shortcomings of current adjuvant strategies. In contrast, the combined inhibition of matrix metalloproteinase and tumour necrosis factor-alpha converting enzyme protected both the cortex and hippocampus in experimental meningitis, and may represent a promising new approach to adjunctive therapy. It is the hope that a more refined molecular understanding of the pathogenesis of brain damage during bacterial meningitis will lead to new adjunctive therapies.


Brain Damage, Chronic/microbiology , Brain Damage, Chronic/pathology , Meningitis, Pneumococcal/pathology , Streptococcus pneumoniae/pathogenicity , Brain Damage, Chronic/enzymology , Brain Damage, Chronic/immunology , Cerebrovascular Circulation , Hippocampus/microbiology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Learning Disabilities/complications , Learning Disabilities/microbiology , Learning Disabilities/pathology , Learning Disabilities/physiopathology , Meningitis, Pneumococcal/complications , Meningitis, Pneumococcal/enzymology , Meningitis, Pneumococcal/immunology , Metalloendopeptidases/metabolism , Reactive Oxygen Species/metabolism , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/physiology
20.
J Heart Lung Transplant ; 20(12): 1310-6, 2001 Dec.
Article En | MEDLINE | ID: mdl-11744415

BACKGROUND: In the early post-operative period after implantation of a continuous flow left ventricular assist device (LVAD) a non-pulsatile flow occurs. We compared the post-operative time-courses of protein S-100B (S100B) and neuron-specific enolase (NSE) as biochemical markers of brain injury in patients after implantation of a continuous flow LVAD and patients receiving a pulsatile flow LVAD. METHODS: Since 1998 the continuous flow DeBakey VAD has been implanted in 8 patients at our institution. For comparison purposes, a group of 7 consecutive patients in whom a pulsatile Novacor N100 LVAD was implanted were investigated. In both groups cardiopulmonary bypass (CPB) with cardiotomy suction was used. S100B and NSE were measured in serum pre-operatively, 4 hours after CPB, and on days 1, 3, 7, and 14 after implantation of the LVAD. A neurologic examination was performed pre-operatively and post-operatively on days 3 and 14. RESULTS: No differences were found between groups in pre-operative characteristics. The analysis of variance with repeated measurements for S-100B and NSE showed significant time effects (p = 0.004, p = 0.009, respectively) but no group effects (p = 0.06, p = 0.26, respectively) and no interaction between groups and time (p = 0.12, p = 0.48, respectively). The pre-operative serum level of S100B was significantly higher (p = 0.03) in the DeBakey VAD group. The pre-operative serum level of NSE was similar in the 2 groups (p = 0.7). In both groups there was a significant increase of S100B and NSE immediately after surgery (S100B: p = 0.006, p = 0.019; NSE: p = 0.01, p = 0.001). The values returned to pre-operative levels in the DeBakey VAD group on day 1 after implantation and in the Novacor group for S100B on day 3 and NSE on day 1. Post-operatively the mean values of S100B and NSE in the DeBakey VAD group compared with the Novacor group were significantly elevated only on day 3 (p = 0.005, p = 0.023).No neurologic complications were noted in patients with a continuous flow LVAD, whereas in the pulsatile LVAD group 2 patients presented neurologic abnormalities during the study period. CONCLUSIONS: The similar course of biochemical markers of brain damage in both groups may indicate that the non-pulsatile flow in the early post-operative period does not lead to increased brain injury or permeability of the brain blood barrier.Elevated levels of S100B and NSE in the post-operative period can be used as diagnostic markers of brain injury in patients after implantation of both types of LVAD.


Brain Damage, Chronic/diagnosis , Calcium-Binding Proteins/blood , Heart-Assist Devices , Nerve Growth Factors/blood , Phosphopyruvate Hydratase/blood , Postoperative Complications/diagnosis , Pulsatile Flow/physiology , S100 Proteins , Adult , Aged , Brain Damage, Chronic/enzymology , Female , Humans , Male , Middle Aged , Postoperative Complications/enzymology , Predictive Value of Tests , Prosthesis Design , S100 Calcium Binding Protein beta Subunit
...