Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(8): e72083, 2013.
Article in English | MEDLINE | ID: mdl-23977213

ABSTRACT

During postnatal development, microglia, the resident innate immune cells of the central nervous system are constantly monitoring the brain parenchyma, cleaning the cell debris, the synaptic contacts overproduced and also maintaining the brain homeostasis. In this context, the postnatal microglia need some control over the innate immune response. One such molecule recently described to be involved in modulation of immune response is TREM2 (triggering receptor expressed on myeloid cells 2). Although some studies have observed TREM2 mRNA in postnatal brain, the regional pattern of the TREM2 protein has not been described. We therefore characterized the distribution of TREM2 protein in mice brain from Postnatal day (P) 1 to 14 by immunostaining. In our study, TREM2 protein was expressed only in microglia/macrophages and is developmentally downregulated in a region-dependent manner. Its expression persisted in white matter, mainly in caudal corpus callosum, and the neurogenic subventricular zone for a longer time than in grey matter. Additionally, the phenotypes of the TREM2+ microglia also differ; expressing CD16/32, MHCII and CD86 (antigen presentation markers) and CD68 (phagocytic marker) in different regions as well as with different intensity till P7. The mannose receptor (CD206) colocalized with TREM2 only at P1-P3 in the subventricular zone and cingulum, while others persisted at low intensities till P7. Furthermore, the spatiotemporal expression pattern and characterization of TREM2 indicate towards its other plausible roles in phagocytosis, progenitor's fate determination or microglia phenotype modulation during postnatal development. Hence, the increase of TREM2 observed in pathologies may recapitulate their function during postnatal development, as a better understanding of this period may open new pathway for future therapies.


Subject(s)
Brain/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Animals , Brain/cytology , Brain/growth & development , Cell Lineage , Gene Expression Regulation, Developmental , Immunity, Innate , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Microglia/metabolism , Organ Specificity , Phenotype , Receptors, Immunologic/genetics
2.
Neurol Res ; 35(3): 308-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23336395

ABSTRACT

BACKGROUND: The zinc finger protein A20 is an ubiquitinating/deubiquitinating enzyme essential for the termination of inflammatory reactions through the inhibition of nuclear factor kappaB (NF-kappaB) signaling. Moreover, it also shows anti-apoptotic activities in some cell types and proapoptotic/pronecrotic effects in others. Although it is known that the regulation of inflammatory and cell death processes are critical in proper brain functioning and that A20 mRNA is expressed in the CNS, its role in the brain under physiological and pathological conditions is still unknown. METHODS: In the present study, we have evaluated the effects of A20 overexpression in mixed cortical cultures in basal conditions: the in vivo pattern of endogenous A20 expression in the control and N-methyl-d-aspartate (NMDA) excitotoxically damaged postnatal day 9 immature rat brain, and the post-injury effects of A20 overexpression in the same lesion model. RESULTS: Our results show that overexpression of A20 in mixed cortical cultures induced significant neuronal death by decreasing neuronal cell counts by 45 ± 9%. in vivo analysis of endogenous A20 expression showed widespread expression in gray matter, mainly in neuronal cells. However, after NMDA-induced excitotoxicity, neuronal A20 was downregulated in the neurodegenerating cortex and striatum at 10-24 hours post-lesion, and it was re-expressed at longer survival times in reactive astrocytes located mainly in the lesion border. When A20 was overexpressed in vivo 2 hours after the excitotoxic damage, the lesion volume at 3 days post-lesion showed a significant increase (20.8 ± 7.0%). No A20-induced changes were observed in the astroglial response to injury. CONCLUSIONS: A20 is found in neuronal cells in normal conditions and is also expressed in astrocytes after brain damage, and its overexpression is neurotoxic for cortical neurons in basal mixed neuron-glia culture conditions and exacerbates postnatal brain excitotoxic damage.


Subject(s)
Brain Diseases/metabolism , Cerebral Cortex/metabolism , DNA-Binding Proteins/biosynthesis , Animals , Astrocytes/metabolism , Brain Diseases/chemically induced , Brain Diseases/pathology , Cells, Cultured , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Immunohistochemistry , N-Methylaspartate/toxicity , NF-kappa B/metabolism , Neurons/metabolism , Rats , Rats, Long-Evans , Rats, Transgenic , Tumor Necrosis Factor alpha-Induced Protein 3 , Up-Regulation
3.
Neurol Res Int ; 2012: 781512, 2012.
Article in English | MEDLINE | ID: mdl-22701792

ABSTRACT

Understanding the evolution of neonatal hypoxic/ischemic is essential for novel neuroprotective approaches. We describe the neuropathology and glial/inflammatory response, from 3 hours to 100 days, after carotid occlusion and hypoxia (8% O(2), 55 minutes) to the C57/BL6 P7 mouse. Massive tissue injury and atrophy in the ipsilateral (IL) hippocampus, corpus callosum, and caudate-putamen are consistently shown. Astrogliosis peaks at 14 days, but glial scar is still evident at day 100. Microgliosis peaks at 3-7 days and decreases by day 14. Both glial responses start at 3 hours in the corpus callosum and hippocampal fissure, to progressively cover the degenerating CA field. Neutrophils increase in the ventricles and hippocampal vasculature, showing also parenchymal extravasation at 7 days. Remarkably, delayed milder atrophy is also seen in the contralateral (CL) hippocampus and corpus callosum, areas showing astrogliosis and microgliosis during the first 72 hours. This detailed and long-term cellular response characterization of the ipsilateral and contralateral hemisphere after H/I may help in the design of better therapeutic strategies.

4.
J Comp Neurol ; 520(12): 2657-75, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22323214

ABSTRACT

The CD200/CD200R inhibitory immune ligand-receptor system regulates microglial activation/quiescence in adult brain. Here, we investigated CD200/CD200R at different stages of postnatal development, when microglial maturation takes place. We characterized the spatiotemporal, cellular, and quantitative expression pattern of CD200 and CD200R in the developing and adult C57/BL6 mice brain by immunofluorescent labeling and Western blotting. CD200 expression increased from postnatal day 1 (P1) to P5-P7, when maximum levels were found, and decreased to adulthood. CD200 was located surrounding neuronal bodies, and very prominently in cortical layer I, where CD200(+) structures included glial fibrillary acidic protein (GFAP)(+) astrocytes until P7. In the hippocampus, CD200 was mainly observed in the hippocampal fissure, where GFAP(+) /CD200(+) astrocytes were also found until P7. CD200(+) endothelium was seen in the hippocampal fissure and cortical blood vessels, notably from P14, showing maximum vascular CD200 in adults. CD200R(+) cells were a population of ameboid/pseudopodic Iba1(+) microglia/macrophages observed at all ages, but significantly decreasing with increasing age. CD200R(+) /Iba1(+) macrophages were prominent in the pial meninges and ventricle lining, mainly at P1-P5. CD200R(+) /Iba1(+) perivascular macrophages were observed in cortical and hippocampal fissure blood vessels, showing maximum density at P7, but being prominent until adulthood. CD200R(+) /Iba1(+) ameboid microglia in the cingulum at P1-P5 were the only CD200R(+) cells in the nervous tissue. In conclusion, the main sites of CD200/CD200R interaction seem to include the molecular layer and pial surface in neonates and blood vessels from P7 until adulthood, highlighting the possible role of the CD200/CD200R system in microglial development and renewal.


Subject(s)
Antigens, CD/metabolism , Brain Chemistry/immunology , Membrane Glycoproteins/metabolism , Neural Inhibition/immunology , Aging/genetics , Aging/immunology , Animals , Animals, Newborn , Antibody Specificity/genetics , Antigen-Antibody Reactions/genetics , Antigens, CD/immunology , Brain Chemistry/genetics , Female , Hippocampus/blood supply , Hippocampus/growth & development , Hippocampus/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Neocortex/blood supply , Neocortex/growth & development , Neocortex/immunology , Neural Inhibition/genetics , Neurogenesis/genetics , Neurogenesis/immunology
5.
J Neurosci Res ; 90(1): 143-59, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21922521

ABSTRACT

Antiinflammatory cytokines such as interleukin-10 (IL-10) have been used to modulate and terminate inflammation and provide neuroprotection. Recently, we reported that the modular recombinant transfection vector NLSCt is an efficient tool for transgene overexpression in vivo, which induces neuroprotection as a result of its RGD-mediated integrin-interacting capacity. We here sought to evaluate the putative synergic neuroprotective action exerted by IL-10 overexpression using NLSCt as a transfection vector after an excitotoxic injury to the postnatal rat brain. For this purpose, lesion volume, neurodegeneration, astroglial and microglial responses, neutrophil infiltration, and proinflammatory cytokine production were analyzed at several survival times after intracortical NMDA injection in postnatal day 9 rats, followed by injection of NLSCt combined with the IL-10 gene, a control transgene, or saline vehicle solution. Our results show no combined neuroprotective effect between RGD-interacting vectors and IL-10 gene therapy; instead, IL-10 overexpression using NLSCt as transfection vector increased lesion volume and neuronal degeneration at 12 hr and 3 days postlesion. In parallel, NLSCt/IL-10 treated animals displayed increased density of neutrophils and microglia/macrophages, and a reduced astroglial content of GFAP and vimentin. Moreover, NLSCt/IL-10 treated animals did not show any variation in interleukin-1ß or tumor necrosis factor-α expression but a slight increase in interleukin-6 content at 7 days postlesion. In conclusion, overexpression of IL-10 by using NLSCt transfection vector did not synergistically neuroprotect the excitotoxically damaged postnatal rat brain but induced changes in the astroglial and microglial and inflammatory cell response.


Subject(s)
Cytokines/metabolism , Interleukin-10/metabolism , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/therapy , Oligopeptides/therapeutic use , Analysis of Variance , Animals , Animals, Newborn , CD11b Antigen/metabolism , Cell Death/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Fluoresceins , Gene Expression Regulation/drug effects , Genetic Vectors/therapeutic use , Glial Fibrillary Acidic Protein/metabolism , Histidine/analogs & derivatives , Histidine/metabolism , Interleukin-10/genetics , Macrophages/drug effects , Macrophages/physiology , Male , N-Methylaspartate/toxicity , Neuroglia/drug effects , Neuroglia/physiology , Neuroprotective Agents/metabolism , Neurotoxicity Syndromes/etiology , Neutrophils/drug effects , Neutrophils/physiology , Oligopeptides/metabolism , Organic Chemicals , Peroxidase/metabolism , Plant Lectins , Rats , Rats, Long-Evans , Transduction, Genetic/methods , Vimentin/metabolism
6.
Exp Gerontol ; 46(9): 723-30, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21601629

ABSTRACT

Brain aging is associated to several morphological and functional alterations that influence the evolution and outcome of CNS damage. Acute brain injury such as an excitotoxic insult induces initial tissue damage followed by associated inflammation and oxidative stress, partly attributed to neutrophil recruitment and the expression of oxidative enzymes such as myeloperoxidase (MPO), among others. However, to date, very few studies have focused on how age can influence neutrophil infiltration after acute brain damage. Therefore, to evaluate the age-dependent pattern of neutrophil cell infiltration following an excitotoxic injury, intrastriatal injection of N-methyl-d-aspartate was performed in young and aged male Wistar rats. Animals were sacrificed at different times between 12h post-lesion (hpl) to 14 days post-lesion (dpl). Cryostat sections were processed for myeloperoxidase (MPO) immunohistochemistry, and double labeling for either neuronal cells (NeuN), astrocytes (GFAP), perivascular macrophages (ED-2), or microglia/macrophages (tomato lectin histochemistry). Our observations showed that MPO + cells were observed in the injured striatum from 12 hpl (when maximum values were found) until 7 dpl, when cell density was strongly diminished. However, at all survival times analyzed, the overall density of MPO + cells was lower in the aged versus the adult injured striatum. MPO + cells were mainly identified as neutrophils (especially at 12 hpl and 1 dpl), but it should be noted that MPO + neurons and microglia/macrophages were also found. MPO + neurons were most commonly observed at 12 hpl and reduced in the aged. MPO + microglia/macrophages were the main population expressing MPO from 3 dpl, when density was also reduced in aged subjects. These results point to neutrophil infiltration as another important factor contributing to the different responses of the adult and aged brain to damage, highlighting the need of using aged animals for the study of acute age-related brain insults.


Subject(s)
Aging/metabolism , Corpus Striatum/metabolism , Corpus Striatum/pathology , Neutrophil Infiltration , Peroxidase/metabolism , Aging/drug effects , Animals , Astrocytes/metabolism , Astrocytes/pathology , Corpus Striatum/drug effects , Excitatory Amino Acid Agonists , Immunohistochemistry , Macrophages/metabolism , Macrophages/pathology , Male , Microglia/metabolism , Microglia/pathology , N-Methylaspartate , Neurons/metabolism , Neurons/pathology , Peroxidase/drug effects , Rats , Rats, Wistar , Time Factors
7.
J Neuropathol Exp Neurol ; 68(4): 391-403, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287312

ABSTRACT

Inflammation is an important determinant of the severity and outcome of central nervous system injury. The endogenous anti-inflammatory cytokine interleukin-10 (IL-10) is upregulated in the injured adult central nervous system where it controls and terminates inflammatory processes. The developing brain, however, displays differences in susceptibility to insults and in associated inflammatory responses from the adult brain; the anatomic and temporal patterns of injury-induced IL-10 expression in the immature brain after excitotoxic injury are unknown. We analyzed the spaciotemporal gene and protein expression of IL-10 and its receptor (IL-10RI) in N-methyl-d-aspartate-induced excitotoxic injury in 9-day-old and control rats using quantitative reverse transcriptase polymerase chain reaction, enzyme-linked immunosorbent assay, and immunohistochemistry. In noninjected control brains, both molecules were expressed mainly in white matter on glial cells and blood vessels; IL-10 was also observed on blood vessels in gray matter and in glial fibrillary acidic protein-positive processes in the hippocampus and near leptomeningeal and ventricle surfaces. In N-methyl-d-aspartate-injected brains, IL-10 gene and protein expression were maximal at 72 hours postinjection; IL-10RI gene and protein expression peaked at 48 hours postinjection. Interleukin-10 and IL-10RI expression in injured areas was mainly found in reactive astrocytes and in microglia/macrophages. The expression patterns of IL-10 and IL-10R suggest possible developmental roles, and their upregulation after injury suggests that this expression may have anti-inflammatory effects in distinct anatomic sites in the immature brain.


Subject(s)
Brain Injuries/pathology , Interleukin-10 Receptor alpha Subunit/metabolism , Interleukin-10/metabolism , Neuroglia/physiology , Up-Regulation/physiology , Animals , Animals, Newborn , Blood Vessels/drug effects , Blood Vessels/metabolism , Brain Injuries/chemically induced , Brain Injuries/physiopathology , Ectodysplasins/metabolism , Excitatory Amino Acid Agonists/toxicity , Female , Glial Fibrillary Acidic Protein/metabolism , Interleukin-10/genetics , Interleukin-10 Receptor alpha Subunit/genetics , Male , N-Methylaspartate/toxicity , Neuroglia/drug effects , Phosphopyruvate Hydratase/metabolism , Plant Lectins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Time Factors , Up-Regulation/drug effects
8.
Clin Immunol ; 128(3): 294-305, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18534912

ABSTRACT

The burden of neurological diseases in western societies has accentuated the need to develop effective therapies to stop the progression of chronic neurological diseases. Recent discoveries regarding the role of the immune system in brain damage coupled with the development of new technologies to manipulate the immune response make immunotherapies an attractive possibility to treat neurological diseases. The wide repertoire of immune responses and the possibility to engineer such responses, as well as their capacity to promote tissue repair, indicates that immunotherapy might offer benefits in the treatment of neurological diseases, similar to the benefits that are being associated with the treatment of cancer and autoimmune diseases. However, before applying such strategies to patients it is necessary to better understand the pathologies to be targeted, as well as how individual subjects may respond to immunotherapies, either in isolation or in combination. Due to the powerful effects of the immune system, one priority is to avoid tissue damage due to the activity of the immune system, particularly considering that the nervous system does not tolerate even the smallest amount of tissue damage.


Subject(s)
Cytokines/immunology , Immunotherapy , Inflammation/immunology , Nervous System Diseases/immunology , Nervous System Diseases/therapy , Alzheimer Disease/therapy , Amyotrophic Lateral Sclerosis/therapy , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Autoimmunity , Cytokines/metabolism , Humans , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mesenchymal Stem Cells/physiology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Nervous System Diseases/metabolism , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/therapy , Parkinson Disease/therapy , Stem Cells/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Mol Cell Neurosci ; 38(2): 170-82, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18434192

ABSTRACT

Mammalian SVZ progenitors continuously generate new neurons in the olfactory bulb. After injury, changes in SVZ cell number suggest injury-induced migration. Studies that trace the migration of SVZ precursors into neurodegenerating areas are lacking. Previously, we showed a decrease in BrdU+SVZ cells following excitotoxic damage to the immature rat cortex. Here, we demonstrate that NMDA-induced injury forces endogenous Cell Tracker Green (CTG) labeled VZ/SVZ precursors out of the SVZ into the neurodegenerating cortex. CTG+/Nestin+/Filamin A+ precursors are closely associated with vimentin+/GFAP+/GLAST+ filaments and express both chemokine receptor CXCR4 and Robo1. In the cortex, SVZ-derived progenitors show a progressive expression of developing, migrating and mature neurons and glial markers. CTG+/GFAP+ astrocytes greatly outnumber CTG+/MAP2+/NeuN+ neurons. SVZ-derived progenitors differentiate into both tbr1+ cortical glutamatergic neurons and calretinin+ interneurons. But, there is little integration of these neurons into the existing circuitry, as seen by Fluorogold retrograde tracing from the internal capsule.


Subject(s)
Cell Movement/physiology , Motor Cortex/pathology , Nerve Degeneration/pathology , Somatosensory Cortex/pathology , Stem Cells/cytology , Age Factors , Animals , Astrocytes/cytology , Biomarkers/metabolism , Calbindin 2 , Cell Differentiation/physiology , Cell Division/physiology , Excitatory Amino Acid Agonists/toxicity , Female , Gliosis/pathology , Internal Capsule/cytology , Internal Capsule/growth & development , Male , Motor Cortex/physiology , N-Methylaspartate/toxicity , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurotoxins/toxicity , Rats , Rats, Long-Evans , Receptors, CXCR4/metabolism , Receptors, Immunologic/metabolism , S100 Calcium Binding Protein G/metabolism , Somatosensory Cortex/physiology , Stem Cells/metabolism , Roundabout Proteins
10.
J Neurosci Res ; 85(16): 3545-56, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17668855

ABSTRACT

Although cleaved caspase-3 is known to be involved in apoptotic cell death mechanisms in neurons, it can also be involved in a nonapoptotic role in astrocytes after postnatal excitotoxic injury. Here we evaluate participation of upstream pathways activating caspase-3 in neurons and glial cells, by studying the intrinsic pathway via caspase-9, the extrinsic pathway via caspase-8, and activation of the p53-dependent pathway. N-methyl-D-aspartate (NMDA) was injected intracortically in 9-day-old postnatal rats, which were sacrificed at several survival times between 4 hr postlesion (pl) and 7 days pl. We analyzed temporal and spatial expression of caspase-8, caspase-9, and p53 and correlation with neuronal and glial markers and caspase-3 activation. Caspase-9 was significantly activated at 10 hpl, strongly correlating with caspase-3. It was present mainly in damaged cortical and hippocampal neurons but was also seen in astrocytes and oligodendrocytes in layer VI and corpus callosum (cc). Caspase-8 showed a diminished correlation with caspase-3. It was present in cortical neurons at 10-72 hpl, showing layer specificity, and also in astroglial and microglial nuclei, mainly in layer VI and cc. p53 Expression increased at 10-72 hpl but did not correlate with caspase-3. p53 Was seen in neurons of the degenerating cortex and in some astrocytes and microglial cells of layer VI and cc. In conclusion, after neonatal excitotoxicity, mainly the mitochondrial intrinsic pathway mediates neuronal caspase-3 and cell death. In astrocytes, caspase-3 is not widely correlated with caspase-8, caspase-9, or p53, except in layer VI-cc astrocytes, where activation of upstream cascades occurs.


Subject(s)
Brain/enzymology , Brain/growth & development , Caspases/metabolism , Nerve Degeneration/enzymology , Neuroglia/enzymology , Neurons/enzymology , Animals , Apoptosis/physiology , Brain/physiopathology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Enzyme Activation/physiology , Female , Gliosis/metabolism , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurotoxins/pharmacology , Rats , Rats, Long-Evans , Signal Transduction/physiology , Time Factors , Tumor Suppressor Protein p53/metabolism
11.
Ann Neurol ; 62(3): 251-61, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17721926

ABSTRACT

OBJECTIVE: Integrin binding to extracellular matrix ligands, including those presenting RGD motifs, modulate diverse cellular processes. In the brain, many endogenous RGD-containing molecules are induced after damage. Previously, the gene therapy vector termed NLSCt, which displays an RGD motif, was shown to neuroprotect after immature brain excitotoxicity. We analyze whether neuroprotection is mediated by the RGD motif. METHODS: RGD-containing synthetic peptide GPenGRGDSPCA (GPen) was injected 2 hours after N-methyl-D-aspartate-mediated excitotoxicity to the postnatal day 9 rat brain. Damage and glial/inflammatory response were evaluated 3 days later. In addition, the neuroprotective effect of GPen and NLSCt after N-methyl-D-aspartate-induced cell death was also analyzed in vitro using neuron-purified and mixed neuron-glia primary cultures. To further characterize whether the neuroprotective effect was mediated by glial-derived soluble factors, we also tested the protective ability of conditioned media from RGD-treated microglia, astrocyte, or mixed glia cultures. RESULTS: Animals treated with GPen peptide showed functional improvement, a significant reduction in lesion volume up to 28%, and a decrease in the number of degenerating neurons. In addition, N-methyl-D-aspartate-injected animals treated with both RGD-containing molecules at the neuroprotective doses showed a significant increase in microglial reactivity and microglia/macrophage cell number, but no differences in neutrophil infiltration and the astroglial response. Finally, in vitro studies showed that the neuroprotective effect was observed in mixed neuron-glia, but not in neuron-purified cultures. Conditioned media from RGD-treated microglial, astroglial, and mixed-glial cultures were not protective. INTERPRETATION: These results suggest that RGD-containing molecules neuroprotect by a glial-dependent mechanism.


Subject(s)
Brain/growth & development , Brain/physiology , Excitatory Amino Acids/toxicity , N-Methylaspartate/toxicity , Neuroglia/physiology , Neuroprotective Agents , Neurotoxicity Syndromes/prevention & control , Oligopeptides/therapeutic use , Animals , Astrocytes/drug effects , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Cell Count , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned , Data Interpretation, Statistical , Densitometry , Female , Immunohistochemistry , Microglia/drug effects , Nerve Degeneration/prevention & control , Neurons/drug effects , Neurons/physiology , Neurotoxicity Syndromes/psychology , Neutrophil Infiltration/drug effects , Pregnancy , Rats , Rats, Long-Evans
12.
Glia ; 55(9): 954-65, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17487878

ABSTRACT

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.


Subject(s)
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
13.
BMC Neurosci ; 7: 35, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16638118

ABSTRACT

BACKGROUND: Superoxide mediated oxidative stress is a key neuropathologic mechanism in acute central nervous system injuries. We have analyzed the neuroprotective efficacy of the transient overexpression of antioxidant enzyme Cu/Zn Superoxide dismutase (SOD) after excitotoxic injury to the immature rat brain by using a recently constructed modular protein vector for non-viral gene delivery termed NLSCt. For this purpose, animals were injected with the NLSCt vector carrying the Cu/Zn SOD or the control GFP transgenes 2 hours after intracortical N-methyl-D-aspartate (NMDA) administration, and daily functional evaluation was performed. Moreover, 3 days after, lesion volume, neuronal degeneration and nitrotyrosine immunoreactivity were evaluated. RESULTS: Overexpression of Cu/Zn SOD transgene after NMDA administration showed improved functional outcome and a reduced lesion volume at 3 days post lesion. In secondary degenerative areas, increased neuronal survival as well as decreased numbers of degenerating neurons and nitrotyrosine immunoreactivity was seen. Interestingly, injection of the NLSCt vector carrying the control GFP transgene also displayed a significant neuroprotective effect but less pronounced. CONCLUSION: When the appropriate levels of Cu/Zn SOD are expressed transiently after injury using the non-viral modular protein vector NLSCt a neuroprotective effect is seen. Thus recombinant modular protein vectors may be suitable for in vivo gene therapy, and Cu/Zn SOD should be considered as an interesting therapeutic transgene.


Subject(s)
Brain Diseases/chemically induced , Brain Diseases/physiopathology , Gene Transfer Techniques , N-Methylaspartate , Neuroprotective Agents , Neurotoxins , Superoxide Dismutase/genetics , Animals , Animals, Newborn , Brain/metabolism , Brain Diseases/pathology , Cell Death/drug effects , Female , Gene Expression , Genetic Vectors/genetics , Male , Neurons/drug effects , Neuroprotective Agents/metabolism , Rats , Rats, Long-Evans , Superoxide Dismutase/metabolism , Transgenes , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis
14.
Neurosci Lett ; 401(1-2): 71-6, 2006 Jun 19.
Article in English | MEDLINE | ID: mdl-16567040

ABSTRACT

Precursor cells have been shown to be affected by oxidative stress, in vivo and vitro, but little is known about the expression of antioxidant mechanisms in neuronal/glial differentiation. We have characterized the expression of Cu/Zn superoxide dismutase (Cu/Zn SOD), one of the main antioxidant proteins involved in the breakdown of superoxide, in the immature rat dorsolateral subventricular zone (SVZ), rostral migratory stream (RMS) and hippocampal subgranular zone (SGZ). Progenitor cells were identified immunohistochemically on cryostat sections by 5'Bromodeoxyuridine (BrdU) incorporation and expressing cells were further characterized using double labeling for progenitor markers. In the SVZ, only a subpopulation of BrdU+ cells, mostly found in the medial SVZ, expressed Cu/Zn SOD. These cells were mostly nestin+ and some were also vimentin+. In contrast, in the lateral SVZ few Cu/Zn SOD+/BrdU+ cells were found. These were primarily nestin+, vimentin-, showed some PSA-NCAM expression, but only a few were NG2+. In the RMS and SGZ virtually all BrdU+ progenitors were Cu/Zn SOD+ and expressed nestin and vimentin. Some RMS cells were also PSA-NCAM+. These findings show a heterogeneous expression of Cu/Zn SOD in restricted cell types in the germinative zones and suggest a role for antioxidant Cu/Zn SOD in progenitor cells of the immature rat brain.


Subject(s)
Antioxidants/metabolism , Brain/growth & development , Neuroglia/enzymology , Neurons/enzymology , Stem Cells/enzymology , Superoxide Dismutase/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/cytology , Brain/enzymology , Bromodeoxyuridine , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation , Intermediate Filament Proteins/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/growth & development , Lateral Ventricles/physiology , Nerve Tissue Proteins/metabolism , Nestin , Neural Cell Adhesion Molecule L1/metabolism , Neuroglia/cytology , Neurons/cytology , Oxidative Stress/physiology , Rats , Rats, Long-Evans , Sialic Acids/metabolism , Vimentin/metabolism
15.
J Neuroinflammation ; 2(1): 12, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15929797

ABSTRACT

BACKGROUND: In the nervous system, as in other organs, Cu/Zn superoxide dismutase (Cu/Zn SOD) is a key antioxidant enzyme involved in superoxide detoxification in normal cellular metabolism and after cell injury. Although it has been suggested that immature brain has a different susceptibility to oxidative damage than adult brain, the distribution and cell-specific expression of this enzyme in immature brain and after postnatal brain damage has not been documented. METHODS: In this study, we used immunohistochemistry and western blot to analyze the expression of Cu/Zn SOD in intact immature rat brain and in immature rat brain after an NMDA-induced excitotoxic cortical injury performed at postnatal day 9. Double immunofluorescence labelling was used to identify Cu/Zn SOD-expressing cell populations. RESULTS: In intact immature brain, Cu/Zn SOD enzyme was widely expressed at high levels in neurons mainly located in cortical layers II, III and V, in the sub-plate, in the pyriform cortex, in the hippocampus, and in the hypothalamus. Glial fibrillary acidic protein-positive cells only showed Cu/Zn SOD expression in the glia limitans and in scattered cells of the ventricle walls. No expression was detected in interfascicular oligodendroglia, microglia or endothelial cells. Following excitotoxic damage, neuronal Cu/Zn SOD was rapidly downregulated (over 2-4 hours) at the injection site before neurodegeneration signals and TUNEL staining were observed. Later, from 1 day post-lesion onward, an upregulation of Cu/Zn SOD was found due to increased expression in astroglia. A further increase was observed at 3, 5 and 7 days that corresponded to extensive induction of Cu/Zn SOD in highly reactive astrocytes and in the astroglial scar. CONCLUSION: We show here that, in the intact immature brain, the expression of Cu/Zn SOD was mainly found in neurons. When damage occurs, a strong and very rapid downregulation of this enzyme precedes neuronal degeneration, and is followed by an upregulation of Cu/Zn SOD in astroglial cells.

16.
BMC Neurosci ; 6: 26, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15826306

ABSTRACT

BACKGROUND: The forebrain subventricular zone (SVZ)-olfactory bulb pathway and hippocampal subgranular zone (SGZ) generate neurons into adulthood in the mammalian brain. Neurogenesis increases after injury to the adult brain, but few studies examine the effect of injury on neural and glial precursors in the postnatal brain. To characterize the spatio-temporal dynamics of cell proliferation in the germinative zones, this study utilized a model of postnatal damage induced by NMDA injection in the right sensorimotor cortex at postnatal day 9. Dividing cell populations were labeled with 5-Bromodeoxyuridine (BrdU) in the intact and damaged postnatal brain. Identity of proliferating cells was determined by double immunolabeling with nestin, GFAP, NeuN and tomato lectin (TL). RESULTS: In the control brain, grouped BrdU+ cells were observed in the Rostral Migratory Stream (RMS), SVZ and SGZ. Maximal proliferation was seen at P12, persisted until P23 and diminished by P49. After injury, a striking reduction in the number of BrdU+ cells was observed in the ipsilateral SVZ from 10 hours (58% decrease) until 14 days post-lesion (88% decrease). In contrast, an increase in grouped BrdU+ cells was seen in the striatum adjacent to the depleted SVZ. Significantly reduced numbers of BrdU+ cells were also seen in the RMS until 3 days post-lesion. No changes were noted in the SGZ. Both in controls and lesioned hemispheres, BrdU+ cells located in the germinal zones were mostly nestin positive and negative for GFAP, NeuN, and TL. In the SVZ area lining the ventricle, BrdU+/nestin+ cells were mainly located between TL+ ependyma and parenchymal GFAP+ astrocytes. After excitotoxicity, a decrease in the number and orientation of GFAP/nestin+ prolongations leaving the SVZ to the cortex, corpus callosum and striatum was noted until 5 days post-lesion. CONCLUSION: Postnatal excitotoxic injury differentially affects proliferating cells in the germinative zones: no change is observed in the dentate gyrus whereas excitotoxicity causes a significant decrease in proliferating cells in the SVZ and RMS. Depletion of BrdU+ cells in the postnatal SVZ and RMS differs from previous studies after adult brain injury and may affect the SVZ-RMS migration and is suggestive of progenitor recruitment to injured areas.


Subject(s)
Brain/cytology , Brain/drug effects , Cell Proliferation/drug effects , Excitatory Amino Acid Agonists/toxicity , Germ Layers/cytology , Germ Layers/drug effects , Animals , Animals, Newborn , Brain/growth & development , Female , Germ Layers/physiology , Male , N-Methylaspartate/toxicity , Rats , Rats, Long-Evans
17.
J Neurotrauma ; 22(1): 189-200, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15665612

ABSTRACT

Oxygen free radicals and nitric oxide (NO) participate in the pathogenesis of acute central nervous system (CNS) injury by forming peroxynitrite, which promotes oxidative damage and tyrosine nitration. Neuronal nitration is associated with cell death, but little is known of the characteristics and cell fate of nitrated astrocytes. In this study, we have used a postnatal excitotoxic lesion model (intracortical NMDA injection) and our aims were (i) to evaluate the temporal and spatial pattern of astroglial nitration in correlation with the neuropathological process and the sources of NO; and (ii) to establish, if any, the correlation among astrocyte nitration and other events such as expression of cytoskeletal proteins, antioxidant enzymes, and cell death markers to cope with nitration and/or undergo cell death. Our results show that after postnatal excitotoxic damage two distinct waves of nitration were observed in relation to astrocytes. At 24 h post-lesion, early-nitrated astrocytes were found within the neurodegenerating area, coinciding with the time of maximal cell death. These early-nitrated astrocytes are highly ramified protoplasmic cells, showing diffuse glial fibrillary acidic protein (GFAP) content and expressing inducible NOS. At later time-points, when astrogliosis is morphologically evident, nitrated hypertrophied reactive astrocytes are observed in the penumbra and the neurodegenerated area, displaying increased expression of GFAP and vimentin cytoskeletal proteins and of metallothionein I-II and Cu/Zn superoxide dismutase antioxidant proteins. Moreover, despite revealing activated caspase-3, they do not show TUNEL labeling. In summary, we show that nitrated astrocytes in vivo constitute a subpopulation of highly reactive astrocytes which display high resistance towards oxidative stress induced cell death.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Animals , Animals, Newborn , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Female , Male , Metallothionein/metabolism , Neurotoxins , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Rats , Rats, Long-Evans , Superoxide Dismutase/metabolism
18.
Stroke ; 33(10): 2499-505, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12364744

ABSTRACT

BACKGROUND AND PURPOSE: The fluorinated salicylate triflusal has been shown to have a neuroprotective effect after an excitotoxic lesion to the postnatal brain. In this regard, the aim of this study was to elucidate whether neuroprotection was associated with changes in the expression of proinflammatory molecules such as interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), inducible nitric oxide synthase (iNOS), or cyclooxygenase-2 (COX-2), well-known mediators of oxidative stress and inflammation, mechanisms underlying secondary damage occurring after excitotoxic/ischemic brain injury. METHODS: Postnatal day 9 rats received an intracortical injection of N-methyl-D-aspartate followed by oral administration of triflusal (30 mg/kg) 8 hours later. Ten or 24 hours after lesion, animals were killed, and brain sections processed for the immunohistochemical demonstration of IL-1beta, TNF-alpha, iNOS, and COX-2. RESULTS: Besides a reduction in the neurodegenerative area, triflusal strongly decreased iNOS immunolabeling at both survival times analyzed, attenuating iNOS immunoreactivity in astroglial cells and infiltrated neutrophils. Additionally, a moderate reduction in COX-2, IL-1beta, and TNF-alpha was observed. Triflusal decreased neuronal and microglial COX-2 expression at 10 and 24 hours after lesion and microglial and astroglial expression of IL-1beta and TNF-alpha at 24 hours after lesion. TNF-alpha expression in neuronal cells at 10 hours after lesion was, however, maintained. CONCLUSIONS: This study suggests that triflusal neuroprotection is associated with a decrease of iNOS and other inflammatory mediators and therefore may constitute a good therapeutic agent in pathological situations in which regulation of inflammatory genes constitutes a relevant step in the outcome of the neurodegenerative event.


Subject(s)
Inflammation Mediators/metabolism , Isoenzymes/metabolism , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/administration & dosage , Nitric Oxide Synthase/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Salicylates/administration & dosage , Administration, Oral , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cyclooxygenase 2 , Disease Models, Animal , Interleukin-1/metabolism , Microglia/drug effects , Microglia/pathology , Microinjections , N-Methylaspartate , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neurotoxins , Neutrophil Infiltration/drug effects , Nitric Oxide Synthase Type II , Rats , Rats, Long-Evans , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Somatosensory Cortex/pathology , Tumor Necrosis Factor-alpha/metabolism
19.
J Neurosci Res ; 68(6): 745-54, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12111835

ABSTRACT

It is well established that after adult brain damage the enzymes cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) play an important role in the inflammatory processes and oxidative stress, which are considered to be the leading factors contributing to delayed cell death. The contribution of these enzymes to postnatal brain damage, however, is poorly understood. In our study, excitotoxic lesions were induced by the injection of N-methyl-D-aspartate in the cortex of postnatal day 9 rats. After different survival times ranging from 4 hr to 7 days post-lesion, brain sections were processed for the immunocytochemical demonstration of COX-2 and iNOS and double labeling with neuronal, glial and neutrophil markers. First and maximal de novo induction of iNOS and COX-2 expression was found at 10 hr post-lesion. Expression of both enzymes started to diminish at 24 hr, reaching basal levels at day 3. iNOS-expressing cells were mainly identified as infiltrated neutrophils as well as highly ramified protoplasmic astrocytes closely associated with blood vessels. Moreover, scattered iNOS-positive neurons were found at the lesion borders. In contrast, COX-2 was mainly observed in reactive microglial cells and neuronal cells. COX-2-positive neurons were found within the degenerating area at 10 hr and at the borders of the lesion later on. This study shows that maximal iNOS and COX-2 expression precedes the period of massive neuronal death observed at 24 hr post-lesion, and may therefore contribute to the evolution of the inflammatory response and the neurodegenerative process after an excitotoxic lesion to the postnatal brain.


Subject(s)
Brain Injuries/enzymology , Cerebral Cortex/enzymology , Isoenzymes/analysis , Nitric Oxide Synthase/analysis , Prostaglandin-Endoperoxide Synthases/analysis , Animals , Brain Injuries/chemically induced , Cell Death , Cerebral Cortex/pathology , Cyclooxygenase 2 , Excitatory Amino Acid Agonists , Immunohistochemistry , N-Methylaspartate , Neurons/enzymology , Nitric Oxide Synthase Type I , Rats , Rats, Long-Evans , Time Factors
20.
J Neurosci Res ; 67(6): 772-80, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11891791

ABSTRACT

NF-kappaB is one of the most important modulators of stress and inflammatory gene expression in the nervous system. In the adult brain, NF-kappaB upregulation has been demonstrated in neurons and glial cells in response to experimental injury and neuropathological disorders, where it has been related to both neurodegenerative and neuroprotective activities. Accordingly, the aim of this study was to evaluate the cellular and temporal patterns of NF-kappaB activation and the expression of its endogenous inhibitor IkappaBalpha following traumatic brain injury (TBI) during the early postnatal weeks, when the brain presents elevated levels of plasticity and neuroprotection. Our results showed that cortical trauma to the 9-day-old rat brain induced a very fast upregulation of NF-kappaB, which was maximal within the first 24 hours after injury. NF-kappaB was mainly observed in neuronal cells of the degenerating cortex as well as in astrocytes located in the corpus callosum adjacent to the injury, where a pulse-like pattern of microglial NF-kappaB activation was also found. In addition, astrocytes of the corpus callosum, and microglial cells to a lower extent, also showed de novo expression of IkappaBalpha within the time of NF-kappaB activation. This study suggests an important role of NF-kappaB activation in the early mechanisms of neuronal death or survival, as well as in the development of the glial and inflammatory responses following traumatic injury to the immature rat brain.


Subject(s)
Brain Injuries/metabolism , Cerebral Cortex/metabolism , DNA-Binding Proteins/biosynthesis , I-kappa B Proteins , NF-kappa B/biosynthesis , Age Factors , Animals , Astrocytes/metabolism , Brain Injuries/pathology , Cerebral Cortex/pathology , Corpus Callosum/metabolism , Corpus Callosum/pathology , DNA-Binding Proteins/analysis , Encephalitis/metabolism , Encephalitis/pathology , Immunohistochemistry , Microglia/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/analysis , Neurons/metabolism , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL
...