Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 54(1): 125-136, 2017 01.
Article in English | MEDLINE | ID: mdl-26732595

ABSTRACT

Methamphetamine (METH)-induced cell death contributes to the pathogenesis of neurotoxicity; however, the relative roles of oxidative stress, apoptosis, and autophagy remain unclear. L-Ascorbate, also called vitamin (Vit.) C, confers partial protection against METH neurotoxicity via induction of heme oxygenase-1. We further investigated the role of Vit. C in METH-induced oxidative stress, apoptosis, and autophagy in cortical cells. Exposure to lower concentrations (0.1, 0.5, 1 mM) of METH had insignificant effects on ROS production, whereas cells exposed to 5 mM METH exhibited ROS production in a time-dependent manner. We confirmed METH-induced apoptosis (by nuclear morphology revealed by Hoechst 33258 staining and Western blot showing the protein levels of pro-caspase 3 and cleaved caspase 3) and autophagy (by Western blot showing the protein levels of Belin-1 and conversion of microtubule-associated light chain (LC)3-I to LC3-II and autophagosome staining by monodansylcadaverine). The apoptosis as revealed by cleaved caspase-3 expression marked an increase at 18 h after METH exposure while both autophagic markers, Beclin 1 and LC3-II, marked an increase in cells exposed to METH for 6 and 24 h, respectively. Treating cells with Vit. C 30 min before METH exposure time-dependently attenuated the production of ROS. Vitamin C also attenuated METH-induced Beclin 1 and LC3-II expression and METH toxicity. Treatment of cells with Vit. C before METH exposure attenuated the expression of cleaved caspase-3 and reduced the number of METH-induced apoptotic cells. We suggest that the protective effect of Vit. C against METH toxicity might be through attenuation of ROS production, autophagy, and apoptosis.


Subject(s)
Apoptosis/drug effects , Ascorbic Acid/pharmacology , Autophagy/drug effects , Cerebral Cortex/drug effects , Methamphetamine/toxicity , Oxidative Stress/drug effects , Animals , Animals, Newborn , Antioxidants/pharmacology , Apoptosis/physiology , Autophagy/physiology , Cells, Cultured , Central Nervous System Stimulants/toxicity , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
2.
Neuropathol Appl Neurobiol ; 42(4): 326-43, 2016 06.
Article in English | MEDLINE | ID: mdl-26245311

ABSTRACT

AIMS: Bacterial meningitis causes high mortality and brain damage. The host immune response is associated with brain injury. Chemokine (C-X-C motif) (CXC) chemokines are neutrophil chemoattractants. This study focused on the beneficial effects of intracerebroventricular administration of reparixin, an inhibitor of chemokine (C-X-C motif) receptor (CXCR)1/2, to rats at 2 h following experimental Klebsiella pneumoniae meningoencephalitis. METHODS: We used a previously established meningoencephalitis animal model in which Sprague-Dawley rats were infected by K. pneumoniae. Sham and infected animals were treated with vehicle or reparixin and sacrificed at various time points. Leukocyte infiltration into cerebrospinal fluid (CSF) and brain as well as gene and protein expression of chemokines and receptors, and neuronal apoptosis were examined. Primary cultures of neuron/glia were infected with K. pneumoniae as an in vitro model of meningoencephalitis. RESULTS: Levels of chemokine (C-X-C motif) ligand (CXCL)2 in CSF time-dependently increased markedly as early as 2 h, and peaked at 8 h following infection and were much higher than those in serum collected simultaneously. Reparixin significantly reduced leukocyte infiltration into CSF and brain tissues, clinical illness, and brain cell apoptosis at 24 h. Reparixin reduced the elevated CSF concentrations of chemokines [CXCL1, CXCL2, chemokine (C-C motif) ligand (CCL)2 and CCL5] and proinflammatory cytokines. Reparixin also reduced the expression of mRNA of various chemokines, chemokine receptors and proinflammatory cytokines in infected brain tissues. Using primary cultures that are devoid of leukocytes, we further observed that reparixin attenuated the neuronal, but not microglial cell death after infection. CONCLUSIONS: Reparixin not only reduces amplified inflammation, but also provides direct neuroprotective effects in K. pneumoniae meningoencephalitis.


Subject(s)
Klebsiella Infections/prevention & control , Meningoencephalitis/microbiology , Meningoencephalitis/prevention & control , Neuroprotective Agents/administration & dosage , Sulfonamides/administration & dosage , Animals , Apoptosis/drug effects , Brain/drug effects , Brain/pathology , Chemokine CXCL2/cerebrospinal fluid , Disease Models, Animal , Inflammation Mediators/cerebrospinal fluid , Klebsiella Infections/complications , Klebsiella Infections/pathology , Male , Meningoencephalitis/complications , Meningoencephalitis/pathology , Neutrophil Infiltration , RNA, Messenger/metabolism , Rats, Sprague-Dawley
3.
J Neuroinflammation ; 12: 147, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26259787

ABSTRACT

BACKGROUND: Neuroinflammation occurs in insulted regions of the brain and may be due to reactive oxygen species (ROS), nitric oxide (NO), cytokines, and chemokines produced by activated glia. Excessive production of neurotoxic molecules causes further neuronal damage. Low levels of vitamin D3 are a risk factor for various brain diseases. METHODS: Using the bacterial endotoxin, lipopolysaccharide (LPS), to induce neuroinflammation in primary cortical neuron-glia cultures, we investigated how 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) affected neuroinflammation. RESULTS: LPS (100 ng/ml) induced the accumulation of nitrite and the production of ROS, interleukin (IL)-6, and macrophage inflammatory protein (MIP)-2 in time-dependent manners. Inhibition of p38 and extracellular signal-regulated kinase (ERK) but not c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK) by 20 µM of SB203580, PD98059, and SP600125, significantly reduced LPS-induced ROS production, NO accumulation, and inducible NO synthase (iNOS) expression, respectively. LPS-induced IL-6 and MIP-2 were significantly attenuated by inhibition of p38, ERK, and JNK MAPK. Cotreatment with 1,25(OH)2D3 attenuated LPS-induced ROS production, NO accumulation, and iNOS expression in concentration-dependent manners. 1,25(OH)2D3 also reduced LPS-induced production of IL-6 and MIP-2. Similarly, iNOS, IL-6, and MIP-2 mRNA expression in cells treated with LPS significantly increased, whereas this effect was attenuated by 1,25(OH)2D3. Moreover, LPS-induced phosphorylation of p38, ERK, and JNK MAPK was significantly inhibited by 1,25(OH)2D3. CONCLUSIONS: Our findings indicate that 1,25(OH)2D3 reduced the LPS-stimulated production of inflammatory molecules in neuron-glia cultures by inhibiting MAPK pathways and the production of downstream inflammatory molecules. We suggest that 1,25(OH)2D3 can be used to alleviate neuroinflammation in various brain injuries.


Subject(s)
Calcitriol/pharmacology , Cerebral Cortex/enzymology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Mitogen-Activated Protein Kinases/metabolism , Neuroglia/enzymology , Neurons/enzymology , Vitamins/pharmacology , Animals , Antioxidants/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Chemokine CXCL2/metabolism , Enzyme Activation/drug effects , Interleukin-6/biosynthesis , MAP Kinase Signaling System/drug effects , Neuroglia/drug effects , Neurons/drug effects , Nitric Oxide Synthase Type II/antagonists & inhibitors , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
4.
PLoS One ; 9(7): e97276, 2014.
Article in English | MEDLINE | ID: mdl-24983461

ABSTRACT

In response to acute insults to the central nervous system, such as pathogen invasion or neuronal injuries, glial cells become activated and secrete inflammatory mediators such as nitric oxide (NO), cytokines, and chemokines. This neuroinflammation plays a crucial role in the pathophysiology of chronic neurodegenerative diseases. Endogenous ascorbate levels are significantly decreased among patients with septic encephalopathy. Using the bacterial endotoxin lipopolysaccharide (LPS) to induce neuroinflammation in primary neuron/glia cocultures, we investigated how L-ascorbate (vitamin C; Vit. C) affected neuroinflammation. LPS (100 ng/ml) induced the expression of inducible NO synthase (iNOS) and the production of NO, interleukin (IL)-6, and macrophage inflammatory protein-2 (MIP-2/CXCL2) in a time-dependent manner; however, cotreatment with Vit. C (5 or 10 mM) attenuated the LPS-induced iNOS expression and production of NO, IL-6, and MIP-2 production. The morphological features revealed after immunocytochemical staining confirmed that Vit. C suppressed LPS-induced astrocytic and microglial activation. Because Vit. C can be transported into neurons and glia via the sodium-dependent Vit. C transporter-2, we examined how Vit. C affected LPS-activated intracellular signaling in neuron/glia cocultures. The results indicated the increased activation (caused by phosphorylation) of mitogen-activated protein kinases (MAPKs), such as p38 at 30 min and extracellular signal-regulated kinases (ERKs) at 180 min after LPS treatment. The inhibition of p38 and ERK MAPK suppressed the LPS-induced production of inflammatory mediators. Vit. C also inhibited the LPS-induced activation of p38 and ERK. Combined treatments of Vit. C and the inhibitors of p38 and ERK yielded no additional inhibition compared with using the inhibitors alone, suggesting that Vit. C functions through the same signaling pathway (i.e., MAPK) as these inhibitors. Vit. C also reduced LPS-induced IκB-α degradation and NF-κB translocation. Thus, Vit. C suppressed the LPS-stimulated production of inflammatory mediators in neuron/glia cocultures by inhibiting the MAPK and NF-κB signaling pathways.


Subject(s)
Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cerebral Cortex/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Cerebral Cortex/pathology , Coculture Techniques , Enzyme Activation/drug effects , Lipopolysaccharides/toxicity , Neuroglia/pathology , Neurons/pathology , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley
5.
J Infect Dis ; 204(10): 1563-72, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21940421

ABSTRACT

There are fewer reports of brain infection by Klebsiella pneumoniae than there are in other organs, but an increase incidence and morbidity has been noted. We have previously developed a rat model of K. pneumoniae meningoencephalitis. Cortistatin (CST) is a recently discovered neuropeptide with endocrine activities in humans. In this study, we found that brain infection by K. pneumoniae increased endogenous prepro-CST messenger RNA expression, which occurred earlier than did leukocyte infiltration in vivo and also occurred in cultured neuron-glia. Postinfection treatment with CST (either intracerebroventricularly or intraperitoneally), but not somatostatin, reduced leukocyte recruitment and clinical illness as revealed by fever and clinical score in vivo. Postinfection increases of proinflammatory cytokine messenger RNA levels were attenuated by CST in neuron-glia cultures, further confirming a direct effect on neuroinflammation. Administration of CST resulted in less postinfection neuronal loss in vitro, suggesting a direct neuroprotective effect and potential as an adjuvant for treating bacterial meningoencephalitis.


Subject(s)
Klebsiella Infections/metabolism , Klebsiella pneumoniae , Meningoencephalitis/metabolism , Neuropeptides/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytokines/metabolism , Disease Models, Animal , Hippocampus/pathology , Klebsiella Infections/drug therapy , Leukocytosis , Male , Meningoencephalitis/drug therapy , Meningoencephalitis/immunology , Necrosis , Neurogenic Inflammation/immunology , Neurogenic Inflammation/metabolism , Neuroimmunomodulation/drug effects , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley
6.
Exp Neurol ; 205(1): 270-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17397834

ABSTRACT

The host immune/inflammatory response following CNS infection by Klebsiella pneumoniae remains poorly understood. Using a rat model of K. pneumoniae meningoencephalitis, we investigated the temporal profiles of brain proinflammatory cytokines and their cellular sources. Leukocyte counts significantly increased in cerebrospinal fluid (CSF) at 2 h after K. pneumoniae inoculation into the rat brain but were still much lower than blood leukocyte counts. However, concentrations of tumor necrosis factor alpha (TNF-alpha), interleukin-1beta (IL-1beta), and IL-6 in CSF were much higher than the simultaneously collected serum levels. The rapid increase in brain expression of these cytokines at the messenger RNA (mRNA) and protein levels occurred earlier than the onset of leukocytosis. Double immunofluorescence staining revealed the presence of TNF-alpha, IL-1beta, and IL-6 in astrocytes and microglia. Exposure of primary culture of glial cells to K. pneumoniae also resulted in time-dependent increases in the concentration of these cytokines in the culture media. Taken together, our results suggest that glial cells are an important early source of proinflammatory cytokines during K. pneumonia infection of CNS.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Klebsiella Infections , Klebsiella pneumoniae , Meningoencephalitis/metabolism , Meningoencephalitis/microbiology , Neuroglia/metabolism , Animals , Brain/metabolism , Culture Media/metabolism , Cytokines/genetics , Humans , Leukocyte Count , Leukocytosis/etiology , Male , Meningoencephalitis/blood , Meningoencephalitis/cerebrospinal fluid , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...