Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neurochem Int ; 61(4): 523-30, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22421531

ABSTRACT

Non-cell autonomous pathology is widely accepted to determine the demise of motoneurons (MNs) in amyotrophic lateral sclerosis (ALS) with astrocytes, GFAP and glutamate transport suggested to play roles in reactive astrogliosis. Previously we described actions of excitotoxicity and oxidative stress to produce differential injury of motoneurons and astrocytes, respectively, and our goal here was to define patterns of MN injury and astrogliosis during a combined excitotoxic-oxidative injury since such a paradigm more closely models disease pathology. Using an in vitro neuronal-glial culture of embryonic mouse spinal cord, we demonstrate that glutamate transport activity was maintained or increased initially, despite a loss of cellular viability, induced by exposure to combinations of excitotoxic [(S)-5-fluorowillardiine (FW)] and oxidative [3-morpholinosydnonimine (SIN-1)] insults over 48 h. Under these conditions, injury was slow in time course and apoptotic-like as shown by the patterns of annexin V and propidium iodide (PI) labelling. Immunocytochemistry for SMI-32 revealed that injury produced time- and insult-dependent reductions in the size of MN arbours, axonal dieback and appreciable neuritic blebbing. These changes were preceded by early hypertrophy of GFAP-positive astrocytes, and followed by more delayed stellation and eventual gliotoxicity. Alterations to EAAT2 immunolabelling were similar to those found for GFAP being initially maintained and then eventually reduced at 48 h. Image analysis of immunocytochemical data confirmed the differential time-dependent changes found with SMI-32, GFAP and EAAT2. Axonopathy and blebbing of MNs was frequently associated with areas of low GFAP immunoreactivity. The exact profile of changes to MNs and astrocytes was context-dependent and sensitive to subtle changes in the mix of excitotoxic-oxidative insults. Overall our findings are consistent with the concepts that the nature, extent and time-course of astrogliosis are insult-dependent, and that discrete pro-survival and destructive components of astrogliosis are likely to determine the precise profile of MN injury in non-cell autonomous pathology of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Axons , Motor Neurons/pathology , Oxidative Stress , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/metabolism , Immunohistochemistry , Mice , Motor Neurons/metabolism
2.
PLoS One ; 7(2): e31176, 2012.
Article in English | MEDLINE | ID: mdl-22363571

ABSTRACT

BACKGROUND: Dorsoventral patterning of the developing spinal cord is important for the correct generation of spinal neuronal types. This process relies in part on cross-repressive interactions between specific transcription factors whose expression is regulated by Sonic hedgehog. Groucho/transducin-like Enhancer of split (TLE) proteins are transcriptional corepressors suggested to be recruited by at least certain Sonic hedgehog-controlled transcription factors to mediate the formation of spatially distinct progenitor domains within the ventral spinal cord. The aim of this study was to characterize the involvement of TLE in mechanisms regulating the establishment of the boundary between the most ventral spinal cord progenitor domains, termed pMN and p3. Because the pMN domain gives rise to somatic motor neurons while the p3 domain generates V3 interneurons, we also examined the involvement of TLE in the acquisition of these neuronal fates. METHODOLOGY AND PRINCIPAL FINDINGS: A combination of in vivo loss- and gain-of-function studies in the developing chick spinal cord was performed to characterize the role of TLE in ventral progenitor domain formation. It is shown here that TLE overexpression causes increased numbers of p3 progenitors and promotes the V3 interneuron fate while suppressing the motor neuron fate. Conversely, dominant-inhibition of TLE increases the numbers of pMN progenitors and postmitotic motor neurons. CONCLUSION: Based on these results, we propose that TLE is important to promote the formation of the p3 domain and subsequent generation of V3 interneurons.


Subject(s)
Interneurons/metabolism , Motor Neurons/metabolism , Neural Stem Cells/metabolism , Repressor Proteins/metabolism , Spinal Cord/cytology , Transcription, Genetic , Animals , Cell Count , Cell Lineage , Chick Embryo , Chickens , Co-Repressor Proteins , Eye Proteins/metabolism , Genes, Dominant/genetics , HEK293 Cells , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Interneurons/cytology , Mice , Mitosis , Models, Biological , Motor Neurons/cytology , Mutant Proteins/metabolism , Neural Stem Cells/cytology , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Transcription Factors/metabolism , Zebrafish Proteins
3.
PLoS One ; 5(11): e13944, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085653

ABSTRACT

BACKGROUND: The ability to precisely identify separate neuronal populations is essential to the understanding of the development and function of different brain structures. This necessity is particularly evident in regions such as the brainstem, where the anatomy is quite complex and little is known about the identity, origin, and function of a number of distinct nuclei due to the lack of specific cellular markers. In this regard, the gene encoding the transcription factor Runx1 has emerged as a specific marker of restricted neuronal populations in the murine central and peripheral nervous systems. The aim of this study was to precisely characterize the expression of Runx1 in the developing and postnatal mouse brainstem. METHODS AND PRINCIPAL FINDINGS: Anatomical and immunohistochemical studies were used to characterize mouse Runx1 expression in the brainstem. It is shown here that Runx1 is expressed in a restricted population of neurons located in the dorsolateral rostral hindbrain. These neurons define a structure that is ventromedial to the dorsal nucleus of the lateral lemniscus, dorsocaudal to the medial paralemniscal nucleus and rostral to the cerebellum. Runx1 expression in these cells is first observed at approximately gestational day 12.5, persists into the adult brain, and is lost in knockout mice lacking the transcription factor Atoh1, an important regulator of the development of neuronal lineages of the rhombic lip. Runx1-expressing neurons in the rostral hindbrain produce cholecystokinin and also co-express members of the Groucho/Transducin-like Enhancer of split protein family. CONCLUSION: Based on the anatomical and molecular characteristics of the Runx1-expressing cells in the rostral hindbrain, we propose that Runx1 expression in this region of the mouse brain defines the superior lateral parabrachial nucleus.


Subject(s)
Brain Stem/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Neurons/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Brain Stem/embryology , Brain Stem/growth & development , Cholecystokinin/genetics , Cholecystokinin/metabolism , Co-Repressor Proteins , Core Binding Factor Alpha 2 Subunit/genetics , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rhombencephalon/embryology , Rhombencephalon/growth & development , Rhombencephalon/metabolism , Time Factors , beta-Galactosidase/metabolism
4.
J Cell Biochem ; 107(6): 1063-72, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19507194

ABSTRACT

Runt-related (RUNX) genes encode evolutionarily conserved transcription factors that play essential roles during development and adult tissue homeostasis. RUNX proteins regulate the transition from proliferation to differentiation in a variety of cell lineages. Moreover, they control the diversification of distinct cellular phenotypes in numerous tissues. Alterations of RUNX functions are associated with several cancers and other human pathologies, underscoring the vital roles of these transcription factors in adult organs. Insights into the functions and regulations of mammalian RUNX proteins have been provided mostly by studies of RUNX involvement in mechanisms of hematopoietic and skeletal development and disease. A growing number of recent investigations are revealing new functions for RUNX family members during the development of the mammalian nervous system. This review will discuss recent progress in the study of RUNX protein involvement in mammalian neural development, with emphasis on the differentiation of olfactory, sensory, and motor neuron lineages.


Subject(s)
Core Binding Factor alpha Subunits/physiology , Neurons/cytology , Stem Cells/cytology , Animals , Cell Lineage , Cell Proliferation , Core Binding Factor alpha Subunits/genetics , Mammals , Mitosis
5.
Glia ; 57(2): 119-35, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18661557

ABSTRACT

In amyotrophic lateral sclerosis (ALS) non-neuronal cells play key roles in disease etiology and loss of motoneurons via noncell-autonomous mechanisms. Reactive astrogliosis and dysfunctional transporters for L-glutamate [excitatory amino acid transporters, (EAATs)] are hallmarks of ALS pathology. Here, we describe mechanistic insights into ALS pathology involving EAAT-associated homeostasis in response to a destructive milieu, in which oxidative stress and excitotoxicity induce respectively astrogliosis and motoneuron injury. Using an in vitro neuronal-glial culture of embryonic mouse spinal cord, we demonstrate that EAAT activity was maintained initially, despite a loss of cellular viability induced by exposure to oxidative [3-morpholinosydnonimine chloride (SIN-1)] and excitotoxic [(S)-5-fluorowillardiine (FW)] conditions. This homeostatic response of EAAT function involved no change in the cell surface expression of EAAT1/2 at 0.5-4 h, but rather alterations in kinetic properties. Over this time-frame, EAAT1/2 both became more widespread across astrocytic arbors in concert with increased expression of glial fibrillary acidic protein (GFAP), although at 8-24 h there was gliotoxicity, especially with SIN-1 rather than FW. An opposite picture was found for motoneurons where FW, not SIN-1, produced early and extensive neuritic shrinkage and blebbing (> or =0.5 h) with somata loss from 2 h. We postulate that EAATs play an early homeostatic and protective role in the pathologic milieu. Moreover, the differential profiles of injury produced by oxidative and excitotoxic insults identify two distinct phases of injury which parallel important aspects of the pathology of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Anterior Horn Cells/metabolism , Astrocytes/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Gliosis/metabolism , Oxidative Stress/physiology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Anterior Horn Cells/pathology , Astrocytes/drug effects , Astrocytes/pathology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Cytoprotection/drug effects , Cytoprotection/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Transporter 2/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis/physiopathology , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Homeostasis/drug effects , Homeostasis/physiology , Mice , Mice, Inbred C57BL , Molsidomine/analogs & derivatives , Molsidomine/toxicity , Neurotoxins/metabolism , Neurotoxins/toxicity , Nitric Oxide Donors/toxicity , Oxidative Stress/drug effects , Time Factors
6.
Neurochem Int ; 48(6-7): 604-10, 2006.
Article in English | MEDLINE | ID: mdl-16530295

ABSTRACT

Excitatory amino acid transporters (EAATs) are responsible for homeostasis of extracellular L-glutamate, and the glial transporters are functionally dominant. EAAT expression or function is altered in acute and chronic neurological conditions, but little is known about the regulation of EAATs in reactive astroglia found in such neuropathologies. These studies examined the effects of the bacterial endotoxin lipopolysaccharide (LPS) on glial EAATs in vitro. The effects of LPS (1 microg/ml, 24-72 h) on EAAT activity and expression were examined in primary cultures of mouse astrocytes. [(3)H]D-aspartate uptake increased to 129% of control by 72 h treatment with LPS. Saturation analysis revealed that apparent K(m) was unchanged whilst V(max) was significantly increased to 172% of control by 72 h LPS treatment. Biotinylation and Western blotting indicated that cell-surface expression of GLT-1 was significantly elevated (146% control) by LPS treatment whereas GLAST expression was unchanged. Confocal analyses revealed that LPS treatment resulted in cytoskeletal changes and stellation of astrocytes, with rearrangement of F-actin (as shown by phalloidin labelling). Immunocytochemistry revealed clustering of GLAST, and increased expression and redistribution of GLT-1 to the cell-surface following treatment with LPS. Similar experiments were conducted in microglia, where LPS (50 ng/ml) was found to up-regulate expression of GLT-1 at 24 and 72 h in concert with cytoskeletal changes accompanying activation. These findings suggest an association of cytoskeletal changes in glia with EAAT activity, with the predominant adaptation involving up-regulation and redistribution of GLT-1.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Lipopolysaccharides/pharmacology , Neuroglia/drug effects , Actins/metabolism , Animals , Animals, Newborn , Aspartic Acid/metabolism , Astrocytes/drug effects , Astrocytes/ultrastructure , Biotinylation , Blotting, Western , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microscopy, Confocal , Neuroglia/cytology , Neuroglia/metabolism , Phenotype , Protein Transport , Up-Regulation
7.
Neurotox Res ; 7(1-2): 143-9, 2005.
Article in English | MEDLINE | ID: mdl-15639805

ABSTRACT

The astrocytic glutamate transporters, EAAT1 and EAAT2, remove released L-glutamate from the synaptic milieu thereby maintaining normal excitatory transmission. EAAT dysfunction during the excitotoxicity and oxidative stress of neurological insults may involve homoeostatic mechanisms associated with astrocytic function. We investigated aspects of EAAT function and expression in concert with astrocytic phenotype in primary cultures of cortical astrocytes and mixed cells of the spinal cord. In spinal cord mixed cultures, hydrogen peroxide (300 microM) reduced both EAAT activity and cellular viability to half of their basal values at 24 h post-treatment, but at 2 h EAAT activity was unaltered, while cellular viability was significantly decreased, suggestive of a mechanism for the maintenance of EAAT activity. Cytochemistry for MAP2, GFAP and propidium iodide revealed that neurons and astrocytes were damaged in a time-dependent manner. A change in astrocyte morphology was observed, with astrocyte cell bodies becoming larger and processes becoming more stellate and often shorter in length. EAAT1 immunoreactivity was reduced at 24 h post-treatment and a re-distribution of the protein was noted after 2 h treatment. In pure astrocytes, lipopolysaccharide (1 microg/ml, 3 d) increased [3H]D-aspartate uptake by 90%, as well EAAT1 immunoreactivity and astrocyte stellation, as shown by immunofluorescent labelling for GFAP. In both culture systems, prominent changes were noted in EAAT function and localization in conjunction with altered astrocytic phenotype. Our findings are indicative of a relationship between astrocytic phenotype and the level of EAAT activity that may be a vital component of astrocytic homeostatic responses in brain injury.


Subject(s)
Astrocytes/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Phenotype , Animals , Astrocytes/drug effects , Cells, Cultured , Hydrogen Peroxide/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/metabolism
8.
J Neurosci Res ; 77(4): 573-82, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15264227

ABSTRACT

Glutamate excitotoxicity is strongly implicated as a major contributing factor in motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Excitotoxicity results from elevated intracellular calcium ion (Ca(2+)) levels, which in turn recruit cell death signaling pathways. Recent evidence suggests that alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor subunit (GluR) stoichiometry is a dominant factor leading to excess Ca(2+) loading in neurodegeneration. In particular, the Ca(2+) permeable glutamate receptor subunit 3 (GluR3) has been implicated in several neurologic conditions such as bipolar disorder and epilepsy. Recent proteomic analysis within our group on the copper zinc superoxide dismutase (SOD1)(G93A) transgenic mouse model of familial ALS (FALS) reveals a potentially deleterious upregulation of GluR3 in spinal cord compared to that in wild-type littermates. Based on this finding we designed a 12mer antisense peptide nucleic acid (PNA) directed against GluR3. This sequence significantly reduced levels of GluR3 protein and protected neuroblastoma x spinal cord (NSC-34) cells against death induced by the AMPA receptor-specific agonist (S)-5-fluorowillardiine. We subsequently treated SOD1(G93A) mice thrice weekly with intraperitoneal injections of the antisense PNA (2.5 mg/kg) commencing at postnatal day 50. Mice treated with the antisense sequence had significantly extended survival compared to mice injected with a nonsense sequence. Western blot analysis, however, did not reveal a significant reduction in GluR3 protein levels in whole extracts of the lumbar spinal cord. These results suggest that interference with the GluR3 component of the AMPA receptor assembly may be a novel strategy for controlling excitotoxic destruction of motor neurons and may lead to new therapeutic opportunities for the treatment of human ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Antisense Elements (Genetics)/pharmacology , Down-Regulation/drug effects , Peptide Nucleic Acids/pharmacology , Receptors, AMPA/antagonists & inhibitors , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Antisense Elements (Genetics)/genetics , Antisense Elements (Genetics)/metabolism , Calcium/metabolism , Calcium/toxicity , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Disease Models, Animal , Disease Progression , Down-Regulation/genetics , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Mice , Mice, Transgenic , Nerve Degeneration/enzymology , Nerve Degeneration/genetics , Nerve Degeneration/therapy , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Superoxide Dismutase/genetics , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...