Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Neurotox Res ; 32(3): 444-459, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28578480

ABSTRACT

Mobile phone communications are conveyed by radiofrequency (RF) electromagnetic fields, including pulse-modulated global system for mobile communications (GSM)-1800 MHz, whose effects on the CNS affected by pathological states remain to be specified. Here, we investigated whether a 2-h head-only exposure to GSM-1800 MHz could impact on a neuroinflammatory reaction triggered by lipopolysaccharide (LPS) in 2-week-old or adult rats. We focused on the cerebral cortex in which the specific absorption rate (SAR) of RF averaged 2.9 W/kg. In developing rats, 24 h after GSM exposure, the levels of cortical interleukin-1ß (IL1ß) or NOX2 NADPH oxidase transcripts were reduced by 50 to 60%, in comparison with sham-exposed animals (SAR = 0), as assessed by RT-qPCR. Adult rats exposed to GSM also showed a 50% reduction in the level of IL1ß mRNA, but they differed from developing rats by the lack of NOX2 gene suppression and by displaying a significant growth response of microglial cell processes imaged in anti-Iba1-stained cortical sections. As neuroinflammation is often associated with changes in excitatory neurotransmission, we evaluated changes in expression and phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the adult cerebral cortex by Western blot analyses. We found that GSM exposure decreased phosphorylation at two residues on the GluA1 AMPAR subunit (serine 831 and 845). The GSM-induced changes in gene expressions, microglia, and GluA1 phosphorylation did not persist 72 h after RF exposure and were not observed in the absence of LPS pretreatment. Together, our data provide evidence that GSM-1800 MHz can modulate CNS cell responses triggered by an acute neuroinflammatory state.


Subject(s)
Cerebral Cortex/growth & development , Cerebral Cortex/immunology , Electromagnetic Fields , Inflammation/metabolism , Neurons/immunology , Acute Disease , Animals , Cell Phone , Cerebral Cortex/pathology , Disease Models, Animal , Inflammation/pathology , Interleukin-1beta/metabolism , Lipopolysaccharides , Male , Microglia/immunology , Microglia/pathology , Microglia/radiation effects , NADPH Oxidase 2/metabolism , Neuroimmunomodulation , Neurons/pathology , RNA, Messenger/metabolism , Rats, Wistar , Receptors, AMPA/metabolism
3.
Glia ; 61(9): 1542-55, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836548

ABSTRACT

The phagocyte NADPH oxidase Nox2 generates superoxide ions implicated in the elimination of microorganisms and the redox control of inflammatory signaling. However, the role of Nox2 in phagocyte functions unrelated to immunity or pathologies is unknown. During development, oriented cell migrations insure the timely recruitment and function of phagocytes in developing tissues. Here, we have addressed the role of Nox2 in the directional migration of microglial cells during development. We show that microglial Nox2 regulates the chemotaxis of purified microglia mediated by the colony stimulating factor-1 receptor (CSF-1R) and the vascular endothelial growth factor receptor-1 (VEGFR1). Stimulation of these receptors triggers activation of Nox2 at the leading edge of polarized cells. In the early postnatal stages of mouse brain development, Nox2 is activated in macrophages / microglial cells in the lateral ventricle or the adjacent subventricular zone (SVZ). Fluorescent microglia injected into the lateral ventricle infiltrate the dorso-caudal SVZ through a mechanism that is blocked by pretreatment of the injected cells with an irreversible Nox inhibitor. Infiltration of endogenous microglia into the caudal SVZ of the cerebral cortex is prevented by (1) Nox2 gene deficiency, (2) treatment with a Nox2 inhibitor (apocynin), and (3) invalidation of the VEGFR1 kinase. We conclude that phagocytes move out of the lateral ventricle soon after birth and infiltrate the cortical SVZ through a mechanism requiring microglial Nox2 and VEGFR1 activation. Nox2 therefore modulates the migration of microglia and their development.


Subject(s)
Chemotaxis/physiology , Green Fluorescent Proteins/metabolism , Lateral Ventricles/cytology , Membrane Glycoproteins/metabolism , Microglia/metabolism , NADPH Oxidases/metabolism , Phagocytes/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Acetophenones/pharmacology , Actins/genetics , Animals , Animals, Newborn , Antigens, Differentiation/metabolism , Bromodeoxyuridine , CD11b Antigen/metabolism , Cell Movement/genetics , Cells, Cultured , Cerebral Cortex/anatomy & histology , Chemotaxis/genetics , Chickens , Enzyme Inhibitors/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor , Green Fluorescent Proteins/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/genetics , Nuclear Proteins/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/genetics
4.
J Neurochem ; 110(5): 1607-16, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19573020

ABSTRACT

We examined the effects of wild-type and mutant atlastin-1 on vesicle transport in the endoplasmic reticulum (ER)-Golgi interface and vesicle budding from ER-derived microsomes using the temperature-sensitive reporter vesicular stomatitis virus glycoprotein (VSV-G), and the ability of purified atlastin-1 to form tubules or vesicles from protein-free phosphatidylserine liposomes. A GTPase domain mutation (T162P) altered the cellular distribution of the ER, but none of the mutations studied significantly affected transport from the ER to the Golgi apparatus. The mutations also had no significant effect on the incorporation of VSV-G into vesicles formed from ER microsomes. Atlastin-1, however, was also incorporated into microsome-derived vesicles, suggesting that it might be implicated in vesicle formation. Purified atlastin-1 transformed phosphatidylserine liposomes into branched tubules and polygonal networks of tubules and vesicles, an action inhibited by GDP and the synthetic dynamin inhibitor dynasore. The GTPase mutations T162P and R217C decreased but did not totally prevent this action; the C-terminal transmembrane domain mutation R495W was as active as the wild-type enzyme. Similar effects were observed in human embryonic kidney cells over-expressing mutant atlastin-1. We concluded that atlastin-1, like dynamin, might be implicated in membrane tubulation and vesiculation and participated in the formation as well as the function of the ER.


Subject(s)
Cytoplasmic Vesicles/enzymology , Endoplasmic Reticulum/enzymology , GTP Phosphohydrolases/metabolism , Membrane Lipids/metabolism , Microtubules/enzymology , Cell Line , Cytoplasmic Vesicles/genetics , Cytoplasmic Vesicles/ultrastructure , Dynamins/genetics , Dynamins/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/ultrastructure , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/physiology , GTP-Binding Proteins , Humans , Membrane Lipids/genetics , Membrane Proteins , Microtubules/genetics , Microtubules/ultrastructure , Protein Transport/physiology , Spastic Paraplegia, Hereditary/enzymology , Spastic Paraplegia, Hereditary/genetics
5.
J Neurosci ; 28(46): 12039-51, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-19005069

ABSTRACT

Reactive oxygen species (ROS) modulate intracellular signaling but are also responsible for neuronal damage in pathological states. Microglia, the resident CNS macrophages, are prominent sources of ROS through expression of the phagocyte oxidase which catalytic subunit Nox2 generates superoxide ion (O2(.-)). Here we show that microglia also express Nox1 and other components of nonphagocyte NADPH oxidases, including p22(phox), NOXO1, NOXA1, and Rac1/2. The subcellular distribution and functions of Nox1 were determined by blocking Nox activity with diphenylene iodonium or apocynin, and by silencing the Nox1 gene in microglia purified from wild-type (WT) or Nox2-KO mice. [Nox1-p22(phox)] dimers localized in intracellular compartments are recruited to phagosome membranes during microglial phagocytosis of zymosan, and Nox1 produces O2(.-) in zymosan-loaded phagosomes. In microglia activated with lipopolysaccharide (LPS), Nox1 produces O2(.-), which enhances cell expression of inducible nitric oxide synthase and secretion of interleukin-1beta. Comparisons of microglia purified from WT, Nox2-KO, or Nox1-KO mice indicate that both Nox1 and Nox2 are required to optimize microglial production of nitric oxide. By injecting LPS in the striatum of WT and Nox1-KO mice, we show that Nox1 also enhances microglial production of cytotoxic nitrite species and promotes loss of presynaptic proteins in striatal neurons. These results demonstrate the functional expression of Nox1 in resident CNS phagocytes, which can promote production of neurotoxic compounds during neuroinflammation. Our study also shows that Nox1- and Nox2-dependent oxidases play distinct roles in microglial activation and that Nox1 is a possible target for the treatment of neuroinflammatory states.


Subject(s)
Encephalitis/enzymology , Gliosis/enzymology , Microglia/enzymology , NADH, NADPH Oxidoreductases/metabolism , Oxidative Stress/physiology , Adaptor Proteins, Signal Transducing , Animals , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Corpus Striatum/physiopathology , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Encephalitis/physiopathology , Female , Gliosis/physiopathology , Inflammation Mediators/pharmacology , Lipopolysaccharides/pharmacology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Neurotoxins/pharmacology , Nitrites/metabolism , Oxidative Stress/drug effects , Proteins/genetics , Proteins/metabolism , Reactive Oxygen Species/metabolism , Zymosan/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
6.
J Neurosci ; 26(12): 3345-56, 2006 Mar 22.
Article in English | MEDLINE | ID: mdl-16554485

ABSTRACT

Because senile plaques in Alzheimer's disease (AD) contain reactive microglia in addition to potentially neurotoxic aggregates of amyloid-beta (Abeta), we examined the influence of microglia on the viability of rodent neurons in culture exposed to aggregated Abeta 1-40. Microglia enhanced the toxicity of Abeta by releasing glutamate through the cystine-glutamate antiporter system Xc-. This may be relevant to Abeta toxicity in AD, because the system Xc(-)-specific xCT gene is expressed not only in cultured microglia but also in reactive microglia within or surrounding amyloid plaques in transgenic mice expressing mutant human amyloid precursor protein or in wild-type mice injected with Abeta. Inhibition of NMDA receptors or system Xc- prevented the microglia-enhanced neurotoxicity of Abeta but also unmasked a neuroprotective effect of microglia mediated by microglial secretion of apolipoprotein E (apoE) in the culture medium. Immunodepletion of apoE or targeted inactivation of the apoE gene in microglia abrogated neuroprotection by microglial conditioned medium, whereas supplementation by human apoE isoforms restored protection, which was potentiated by the presence of microglia-derived cofactors. These results suggest that inhibition of microglial system Xc- might be of therapeutic value in the treatment of AD. Its inhibition not only prevents glutamate excitotoxicity but also facilitates neuroprotection by apoE.


Subject(s)
Alzheimer Disease/metabolism , Amino Acid Transport System y+/metabolism , Amyloid beta-Peptides/toxicity , Apolipoproteins E/metabolism , Microglia/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Peptide Fragments/toxicity , Alzheimer Disease/physiopathology , Amino Acid Transport System y+/genetics , Animals , Apolipoproteins E/genetics , Cell Communication/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Wistar
7.
Neuron ; 41(4): 535-47, 2004 Feb 19.
Article in English | MEDLINE | ID: mdl-14980203

ABSTRACT

The loss of neuronal cells, a prominent event in the development of the nervous system, involves regulated triggering of programmed cell death, followed by efficient removal of cell corpses. Professional phagocytes, such as microglia, contribute to the elimination of dead cells. Here we provide evidence that, in addition to their phagocytic activity, microglia promote the death of developing neurons engaged in synaptogenesis. In the developing mouse cerebellum, Purkinje cells die, and 60% of these neurons that already expressed activated caspase-3 were engulfed or contacted by spreading processes emitted by microglial cells. Apoptosis of Purkinje cells in cerebellar slices was strongly reduced by selective elimination of microglia. Superoxide ions produced by microglial respiratory bursts played a major role in this Purkinje cell death. Our study illustrates a mammalian form of engulfment-promoted cell death that links the execution of neuron death to the scavenging of dead cells.


Subject(s)
Apoptosis/physiology , Cell Communication/physiology , Cerebellar Cortex/growth & development , Microglia/physiology , Purkinje Cells/physiology , Signal Transduction/physiology , Animals , Antibodies/pharmacology , Caspase 3 , Caspases/metabolism , Cell Differentiation/physiology , Cell Respiration/drug effects , Cell Respiration/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cerebellar Cortex/cytology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Presynaptic Terminals/physiology , Purkinje Cells/cytology , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...