Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Neuroimmunomodulation ; 29(4): 425-432, 2022.
Article in English | MEDLINE | ID: mdl-35705003

ABSTRACT

BACKGROUND: Ischemic stroke is a major health issue that causes high incidents of morbidity and mortality worldwide. Irisin is an excise-induced protein that has exhibited pleiotropic properties. Accumulating evidence reveals its critical roles in the regulation of various cellular functions, including nervous system functions. This study aims to disclose the effect of irisin on rat cerebral neurons suffering from hypoxia/reoxygenation (H/R) treatment and to explore the potential underlying molecular mechanisms. METHODS: The percentage of rat cerebral neuron cell death was determined by flow cytometry analysis and MTT assay. The expression levels of target genes were measured by western blotting and real-time quantitative reverse transcription PCR assay. RESULTS: Our results demonstrated that irisin treatment substantially reduced H/R-induced apoptosis of rat cerebral neurons. Further investigation revealed that irisin treatment markedly decreased mitogen-activated protein kinase (MAPK) signaling pathway activation and suppressed pro-informatory cytokine expression in cerebral neurons with H/R challenge. Finally, we showed that the neuroprotective effect and anti-inflammatory effect of irisin were comparable with three MAPK signaling inhibitors. CONCLUSION: Irisin exerts profound neuroprotective and anti-inflammatory effects on H/R-stimulated cerebral neurons by inhibiting the MAPK signaling activation. Therefore, irisin may serve as a potential drug for the treatment of patients with ischemic stroke.


Subject(s)
Fibronectins , Ischemic Stroke , Animals , Rats , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/pharmacology , Apoptosis/genetics , Apoptosis/immunology , Cytokines/genetics , Cytokines/immunology , Fibronectins/genetics , Fibronectins/immunology , Fibronectins/pharmacology , Hypoxia, Brain/genetics , Hypoxia, Brain/immunology , Ischemic Stroke/genetics , Ischemic Stroke/immunology , Neurons/immunology
2.
Sci Rep ; 10(1): 6449, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32296107

ABSTRACT

Hypoxia (HYPX) induced-overload Ca2+ entry results in increase of mitochondrial oxidative stress, inflammation and apoptosis in several neurons. Ca2+ permeable TRPM2 channel was gated by ADP-ribose (ADPR) and reactive oxygen species (ROS), although its activity was modulated in HYPX-exposed neurons by resveratrol (RSV). The aim of this study was to evaluate if a therapy of RSV can modulate the effect of HYPX in the TRPM2 expressing SH-SY5Y neuronal and HEK293 (no expression of TRPM2) cell lines. The SH-SY5Y and HEK293 cells were divided into four groups as control, RSV (50 µM and 24 hours), and HYPX and RSV + HYPX. For induction of HYPX in the cells, CoCl2 (200 µM and 24 hours) incubation was used. HYPX-induced intracellular Ca2+ responses to TRPM2 activation were increased in the SH-SY5Y cells but not in the HEK293 cells from coming H2O2 and ADPR. RSV treatment improved intracellular Ca2+ responses, mitochondrial function, suppressed the generation of cytokine (IL-1ß and TNF-α), cytosolic and mitochondrial ROS in the SH-SY5Y cells. Intracellular free Zn2+, apoptosis, cell death, PARP-1, TRPM2 expression, caspase -3 and -9 levels are increased through activating TRPM2 in the SH-SY5Y cells exposed to the HYPX. However, the values were decreased in the cells by RSV and TRPM2 blockers (ACA and 2-APB). In SH-SY5Y neuronal cells exposed to HYPX conditions, the neuroprotective effects of RSV were shown to be exerted via modulation of oxidative stress, inflammation, apoptosis and death through modulation of TRPM2 channel. RSV could be used as an effective agent in the treatment of neurodegeneration exposure to HYPX.


Subject(s)
Apoptosis/drug effects , Mitochondria/drug effects , Neurons/drug effects , Resveratrol/pharmacology , TRPM Cation Channels/metabolism , Adenosine Diphosphate Ribose/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/immunology , Cell Line, Tumor , Cobalt/toxicity , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hypoxia, Brain/drug therapy , Hypoxia, Brain/immunology , Hypoxia, Brain/pathology , Mitochondria/metabolism , Mitochondria/pathology , Neurons/cytology , Neurons/immunology , Neurons/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Resveratrol/therapeutic use
3.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R611-R622, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351418

ABSTRACT

Moderate anemia is associated with increased mortality and morbidity, including acute kidney injury (AKI), in surgical patients. A red blood cell (RBC)-specific antibody model was utilized to determine whether moderate subacute anemia could result in tissue hypoxia as a potential mechanism of injury. Cardiovascular and hypoxic cellular responses were measured in transgenic mice capable of expressing hypoxia-inducible factor-1α (HIF-1α)/luciferase activity in vivo. Antibody-mediated anemia was associated with mild intravascular hemolysis (6 h) and splenic RBC sequestration ( day 4), resulting in a nadir hemoglobin concentration of 89 ± 13 g/l on day 4. At this time point, renal tissue oxygen tension (PtO2) was decreased in anemic mice relative to controls (13.1 ± 4.3 vs. 20.8 ± 3.7 mmHg, P < 0.001). Renal tissue hypoxia was associated with an increase in HIF/luciferase expression in vivo ( P = 0.04) and a 20-fold relative increase in renal erythropoietin mRNA transcription ( P < 0.001) but no increase in renal blood flow ( P = 0.67). By contrast, brain PtO2 was maintained in anemic mice relative to controls (22.7 ± 5.2 vs. 23.4 ± 9.8 mmHg, P = 0.59) in part because of an increase in internal carotid artery blood flow (80%, P < 0.001) and preserved cerebrovascular reactivity. Despite these adaptive changes, an increase in brain HIF-dependent mRNA levels was observed (erythropoietin: P < 0.001; heme oxygenase-1: P = 0.01), providing evidence for subtle cerebral tissue hypoxia in anemic mice. These data demonstrate that moderate subacute anemia causes significant renal tissue hypoxia, whereas adaptive cerebrovascular responses limit the degree of cerebral tissue hypoxia. Further studies are required to assess whether hypoxia is a mechanism for acute kidney injury associated with anemia.


Subject(s)
Acute Kidney Injury/blood , Anemia/blood , Antibodies, Monoclonal , Brain/blood supply , Erythrocytes/metabolism , Hypoxia, Brain/blood , Kidney/blood supply , Oxygen/blood , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Anemia/immunology , Anemia/pathology , Anemia/physiopathology , Animals , Brain/metabolism , Brain/pathology , Cerebrovascular Circulation , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/pathology , Erythropoietin/genetics , Erythropoietin/metabolism , Glycophorins/blood , Glycophorins/immunology , Hemolysis , Hypoxia, Brain/immunology , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice, Transgenic , Renal Circulation , Severity of Illness Index , Spleen/metabolism , Spleen/pathology , Up-Regulation
4.
Cell Mol Immunol ; 14(8): 693-701, 2017 Aug.
Article in English | MEDLINE | ID: mdl-26707403

ABSTRACT

Hypoxic-ischemic brain damage (HIBD) is a common cause of infant death. The purpose of our research was to explore the immunoregulatory mechanism of placenta-derived mesenchymal stem cells (PD-MSCs) in HIBD treatment. Seven-day-old rat pups were randomly divided into HIBD, PD-MSC, fibroblast, and control groups. Forty-eight hours after HIBD induction, cells at a density of 5 × 104 cells/10 µl were injected into the cerebral tissue in the PD-MSC and fibroblast groups. The TNF-α, interleukin- 17 (IL-17), interferon-γ (IFN-γ), and IL-10 levels were detected through quantitative real-time polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Regulatory T cell (Tregs) populations were detected through flow cytometry, and forkhead box P3 (Foxp3) was measured through western blot analysis. Behavioral tests and gross and pathological examinations showed that PD-MSC treatment exerted significantly stronger neuroprotective effects than the other treatments. The expression levels of pro-inflammatory cytokines were substantially upregulated after HI injury. Compared with fibroblast treatment, PD-MSC treatment inhibited the production of pro-inflammatory cytokines and increased the production of IL-10 in the ischemic hemispheres and peripheral blood serum (all P < 0.01). Flow cytometry results showed a notable increase in the number of Tregs within the spleen of the HIBD group. Moreover, the number of Tregs and the Foxp3 expression levels were higher in the PD-MSC treatment group than in the HIBD and fibroblast groups (all P < 0.01). Our research suggests that the mechanism of PD-MSC treatment for HIBD partially involves inflammatory response suppression.


Subject(s)
Hypoxia, Brain/therapy , Hypoxia/therapy , Inflammation/therapy , Ischemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Placenta/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Animals, Newborn , Behavior Rating Scale , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Hypoxia/immunology , Hypoxia, Brain/immunology , Immunosuppression Therapy , Infant , Inflammation/immunology , Ischemia/immunology , Neuroprotection , Pregnancy , Rats
5.
Endocr Rev ; 37(4): 372-402, 2016 08.
Article in English | MEDLINE | ID: mdl-27196727

ABSTRACT

Inflammatory activation of microglia is a hallmark of several disorders of the central nervous system. In addition to protecting the brain against inflammatory insults, microglia are neuroprotective and play a significant role in maintaining neuronal connectivity, but the prolongation of an inflammatory status may limit the beneficial functions of these immune cells. The finding that estrogen receptors are present in monocyte-derived cells and that estrogens prevent and control the inflammatory response raise the question of the role that this sex steroid plays in the manifestation and progression of pathologies that have a clear sex difference in prevalence, such as multiple sclerosis, Parkinson's disease, and Alzheimer's disease. The present review aims to provide a critical review of the current literature on the actions of estrogen in microglia and on the involvement of estrogen receptors in the manifestation of selected neurological disorders. This current understanding highlights a research area that should be expanded to identify appropriate replacement therapies to slow the progression of such diseases.


Subject(s)
Demyelinating Diseases , Estrogens/metabolism , Hypoxia, Brain , Inflammation , Microglia , Neurodegenerative Diseases , Receptors, Estrogen/metabolism , Stroke , Animals , Demyelinating Diseases/immunology , Demyelinating Diseases/metabolism , Humans , Hypoxia, Brain/immunology , Hypoxia, Brain/metabolism , Inflammation/immunology , Inflammation/metabolism , Microglia/immunology , Microglia/metabolism , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Stroke/immunology , Stroke/metabolism
6.
Physiol Rep ; 4(6)2016 Mar.
Article in English | MEDLINE | ID: mdl-27033443

ABSTRACT

Transient hypoxia in pregnancy stimulates a physiological reflex response that redistributes blood flow and defends oxygen delivery to the fetal brain. We designed the present experiment to test the hypotheses that transient hypoxia produces damage of the cerebral cortex and that ketamine, an antagonist ofNMDAreceptors and a known anti-inflammatory agent, reduces the damage. Late gestation, chronically catheterized fetal sheep were subjected to a 30-min period of ventilatory hypoxia that decreased fetal PaO2from 17 ± 1 to 10 ± 1 mmHg, or normoxia (PaO217 ± 1 mmHg), with or without pretreatment (10 min before hypoxia/normoxia) with ketamine (3 mg/kg, i.v.). One day (24 h) after hypoxia/normoxia, fetal cerebral cortex was removed andmRNAextracted for transcriptomics and systems biology analysis (n = 3-5 per group). Hypoxia stimulated a transcriptomic response consistent with a reduction in cellular metabolism and an increase in inflammation. Ketamine pretreatment reduced both of these responses. The inflammation response modeled with transcriptomic systems biology was validated by immunohistochemistry and showed increased abundance of microglia/macrophages after hypoxia in the cerebral cortical tissue that ketamine significantly reduced. We conclude that transient hypoxia produces inflammation of the fetal cerebral cortex and that ketamine, in a standard clinical dose, reduces the inflammation response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cerebral Cortex/drug effects , Fetal Hypoxia/drug therapy , Hypoxia, Brain/drug therapy , Inflammation Mediators/metabolism , Ketamine/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Female , Fetal Hypoxia/genetics , Fetal Hypoxia/immunology , Fetal Hypoxia/metabolism , Fetal Hypoxia/pathology , Gene Expression Profiling/methods , Gene Expression Regulation , Gestational Age , Hypoxia, Brain/genetics , Hypoxia, Brain/immunology , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Inflammation Mediators/immunology , Oligonucleotide Array Sequence Analysis , Pregnancy , RNA, Messenger/metabolism , Sheep , Systems Biology , Time Factors
7.
Purinergic Signal ; 12(1): 89-101, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26608888

ABSTRACT

Neonatal brain hypoxic ischemia (HI) often results in long-term motor and cognitive impairments. Post-ischemic inflammation greatly effects outcome and adenosine receptor signaling modulates both HI and immune cell function. Here, we investigated the influence of adenosine A1 receptor deficiency (A1R(-/-)) on key immune cell populations in a neonatal brain HI model. Ten-day-old mice were subjected to HI. Functional outcome was assessed by open locomotion and beam walking test and infarction size evaluated. Flow cytometry was performed on brain-infiltrating cells, and semi-automated analysis of flow cytometric data was applied. A1R(-/-) mice displayed larger infarctions (+33%, p < 0.05) and performed worse in beam walking tests (44% more mistakes, p < 0.05) than wild-type (WT) mice. Myeloid cell activation after injury was enhanced in A1R(-/-) versus WT brains. Activated B lymphocytes expressing IL-10 infiltrated the brain after HI in WT, but were less activated and did not increase in relative frequency in A1R(-/-). Also, A1R(-/-) B lymphocytes expressed less IL-10 than their WT counterparts, the A1R antagonist DPCPX decreased IL-10 expression whereas the A1R agonist CPA increased it. CD4(+) T lymphocytes including FoxP3(+) T regulatory cells, were unaffected by genotype, whereas CD8(+) T lymphocyte responses were smaller in A1R(-/-) mice. Using PCA to characterize the immune profile, we could discriminate the A1R(-/-) and WT genotypes as well as sham operated from HI-subjected animals. We conclude that A1R signaling modulates IL-10 expression by immune cells, influences the activation of these cells in vivo, and affects outcome after HI.


Subject(s)
Brain Ischemia/immunology , Brain/immunology , Hypoxia, Brain/immunology , Receptor, Adenosine A1/immunology , Adenosine A1 Receptor Agonists/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cerebral Infarction/pathology , Female , Hypoxia, Brain/congenital , Interleukin-10/biosynthesis , Interleukin-10/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Postural Balance , Pregnancy , Receptor, Adenosine A1/drug effects
8.
J Matern Fetal Neonatal Med ; 29(2): 309-16, 2016.
Article in English | MEDLINE | ID: mdl-25655529

ABSTRACT

AIM: Circulating immune cell activation is associated with worse outcome in adult and animal models of brain injury. Our aim was to profile the systemic inflammatory response over the first week of life in infants at risk of neonatal encephalopathy (NE) and correlate early neutrophil and monocyte endotoxin and activation responses with outcome. METHODS: Prospective observational study in a tertiary referral university hospital including 22 infants requiring resuscitation at birth who had serial (five time points) neutrophil and monocyte CD11b (marker of cell adhesion), intracellular reactive oxygen intermediates (ROI; cell activation) and Toll-like receptor (TLR; endotoxin recognition) before and after endotoxin stimulation ex vivo compared to neonatal controls. RESULTS: All neonates requiring resuscitation at delivery (n = 122 samples) had higher neutrophil and monocyte CD11b and TLR-4 expression compared with adults and neonatal controls. Neonates with abnormal neuroimaging and/or severe NE had increased CD11b, ROI and TLR-4. Increased polymorphonuclear leukocytes TLR-4 expression was associated with increased mortality in infants with NE. CONCLUSION: Innate immune dysregulation in the first week of life is associated with severity of outcome in neonatal brain injury in this cohort and may be amenable to immunomodulation.


Subject(s)
CD11b Antigen/metabolism , Hypoxia, Brain/immunology , Phagocytes/metabolism , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism , Adult , Female , Humans , Infant, Newborn , Male , Neutrophil Activation , Prospective Studies , Retrospective Studies
10.
J Neurochem ; 126(4): 437-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23713463

ABSTRACT

Guanosine (GUO) is an endogenous modulator of glutamatergic excitotoxicity and has been shown to promote neuroprotection in in vivo and in vitro models of neurotoxicity. This study was designed to understand the neuroprotective mechanism of GUO against oxidative damage promoted by oxygen/glucose deprivation and reoxygenation (OGD). GUO (100 µM) reduced reactive oxygen species production and prevented mitochondrial membrane depolarization induced by OGD. GUO also exhibited anti-inflammatory actions as inhibition of nuclear factor kappa B activation and reduction of inducible nitric oxide synthase induction induced by OGD. These GUO neuroprotective effects were mediated by adenosine A1 receptor, phosphatidylinositol-3 kinase and MAPK/ERK. Furthermore, GUO recovered the impairment of glutamate uptake caused by OGD, an effect that occurred via a Pertussis toxin-sensitive G-protein-coupled signaling, blockade of adenosine A2A receptors (A2A R), but not via A1 receptor. The modulation of glutamate uptake by GUO also involved MAPK/ERK activation. In conclusion, GUO, by modulating adenosine receptor function and activating MAPK/ERK, affords neuroprotection of hippocampal slices subjected to OGD by a mechanism that implicates the following: (i) prevention of mitochondrial membrane depolarization, (ii) reduction of oxidative stress, (iii) regulation of inflammation by inhibition of nuclear factor kappa B and inducible nitric oxide synthase, and (iv) promoting glutamate uptake.


Subject(s)
Encephalitis , Guanosine/metabolism , Hippocampus/immunology , Hippocampus/metabolism , Hypoxia, Brain , Animals , Cell Survival/drug effects , Cell Survival/physiology , Encephalitis/drug therapy , Encephalitis/immunology , Encephalitis/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Glucose/pharmacology , Glutamic Acid/pharmacokinetics , Guanosine/pharmacology , Hippocampus/cytology , Hypoxia, Brain/drug therapy , Hypoxia, Brain/immunology , Hypoxia, Brain/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Membrane Potential, Mitochondrial/physiology , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Organ Culture Techniques , Oxidative Stress/drug effects , Oxidative Stress/physiology , Oxygen/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor, Adenosine A1/metabolism , Synaptotagmins , Tritium
11.
Georgian Med News ; (203): 17-22, 2012 Feb.
Article in Russian | MEDLINE | ID: mdl-22466535

ABSTRACT

In this article there are materials, which were received in the process of treatment of 69 patients with azaleptin (leponeks) acute severe poisonings. The immunologic research has shown that the first phase of acute poisonings in patients with toxicohypoxic encephalopathy is accompanied by severe immune alterations. The alterations of proinflammation and antiinflammation cytokines have been investigated and its increase has not been revealed. It was shown, that the employment of citoflavin in intensive therapy of acute poisonings leads to decrease of the level of immunosupression.


Subject(s)
Flavin Mononucleotide/administration & dosage , Hypoxia, Brain/pathology , Immune System/pathology , Inosine Diphosphate/administration & dosage , Neurotoxicity Syndromes/pathology , Niacinamide/administration & dosage , Poisoning/therapy , Succinates/administration & dosage , Blood Cell Count , Clozapine/therapeutic use , Critical Illness/therapy , Drug Combinations , Female , Humans , Hypoxia, Brain/blood , Hypoxia, Brain/chemically induced , Hypoxia, Brain/immunology , Male , Neurotoxicity Syndromes/blood , Neurotoxicity Syndromes/immunology
12.
Exp Neurol ; 232(2): 329-32, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21945008

ABSTRACT

The aim of this study was to investigate the comparative effects of transforming growth factor ß1 (TGF-ß1) and interleukin-10 (IL-10) on the repeated brief hypoxia-induced alterations in the activity of hippocampal slice CA1 pyramidal neurons. The method of field potentials measurement in CA1 region of hippocampal slices was used. The principal results of our work are summarized as follow. 1. TGF-ß1 reduces the depressive effect of brief hypoxia on the population spike amplitude more effectively than IL-10. 2. During TGF-ß1 exposure (in contrast to IL-10), three 3-min hypoxic episodes do not induce the rapid hypoxic preconditioning. 3. TGF-ß1 and IL-10 equally abolish posthypoxic hyperexcitability induced by repeated brief episodes of hypoxia in CA1 pyramidal neurons. These findings indicated that TGF-ß1 and IL-10 are able to evoke anti-hypoxic effect and abolish the development of posthypoxic hyperexcitability induced by repeated brief hypoxic episodes in hippocampal CA1 pyramidal neurons. Our results also demonstrated that TGF-ß1 reduced the effectiveness of hypoxia to depress neuronal activity more effectively than IL-10. We suggest that the present findings allow to explain the certain neuroprotective mechanisms of IL-10 and TGF-beta1 in the early phase of hypoxia and indicate that a therapeutic anti-inflammatory approach using these substances can provide neuroprotection in the brain hypoxic conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CA1 Region, Hippocampal , Hypoxia, Brain , Interleukin-10/pharmacology , Transforming Growth Factor beta1/pharmacology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/immunology , CA1 Region, Hippocampal/physiopathology , Hypoxia, Brain/drug therapy , Hypoxia, Brain/immunology , Hypoxia, Brain/physiopathology , Ischemic Preconditioning/methods , Male , Membrane Potentials/drug effects , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Wistar
13.
Naunyn Schmiedebergs Arch Pharmacol ; 384(2): 177-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21713382

ABSTRACT

R(-) enantiomers of the 2-arylpropionic acid derivatives ibuprofen and flurbiprofen weakly inhibit cyclooxygenase (COX) activity. However, a possible cytoprotective effect has been proposed. The aim of the study is to investigate the possible mechanism of this effect. An in vitro hypoxia-reoxygenation model in rat brain slices was used (n=6 rats per group). After reoxygenation, we measured LDH efflux (neuronal death), brain prostaglandin E(2) (PGE(2)) concentration, interleukins (IL)-1ß and 10, oxidative and nitrosative stress (lipid peroxides, glutathione, 3-nitrotyrosine, and nitrites/nitrates). Anti-COX activity was measured in human whole blood. Racemic, R(-), and S(+) enantiomers of ibuprofen and flurbiprofen were tested. All compounds had a cytoprotective effect with IC(50) values in the range of 10(-5) M. R(-) enantiomers did not significantly inhibit brain PGE(2). The concentration of IL-1ß was reduced by 53.1% by the racemic form, 30.6% by the S(+) and 43.2% by the R(-) enantiomer of ibuprofen. The IL-10 concentration increased significantly only with S(+)-flurbiprofen (33.1%) and R(-)-flurbiprofen (26.1%). Lipid peroxidation was significantly reduced by all three forms of flurbiprofen. Nitrite + nitrate concentrations were reduced by racemic, S(+), and R(-)-flurbiprofen. Peroxynitrite formation (3-nitrotyrosine) was significantly reduced by racemic and S(+)-ibuprofen. COX inhibition is not the main mechanism of cytoprotection for these compounds. Their influence on inflammatory mediators and oxidative and nitrosative stress could account for the potential cytoprotective effect of R(-) enantiomers.


Subject(s)
Brain/drug effects , Cyclooxygenase Inhibitors/pharmacology , Cytoprotection/drug effects , Flurbiprofen/pharmacology , Ibuprofen/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Brain/enzymology , Brain/immunology , Brain/pathology , Cyclooxygenase Inhibitors/chemistry , Cytokines/metabolism , Dinoprostone/metabolism , Flurbiprofen/chemistry , Hypoxia, Brain/enzymology , Hypoxia, Brain/immunology , Hypoxia, Brain/pathology , Hypoxia, Brain/prevention & control , Ibuprofen/chemistry , In Vitro Techniques , Lipid Peroxidation/drug effects , Male , Rats , Rats, Wistar , Stereoisomerism , Tyrosine/analogs & derivatives , Tyrosine/analysis
14.
Front Biosci (Schol Ed) ; 3(3): 884-900, 2011 06 01.
Article in English | MEDLINE | ID: mdl-21622239

ABSTRACT

The developing brain is susceptible to hypoxic damage because of its high oxygen and energy requirements. Hypoxia-induced inflammatory response has been recognized as one of the main culprits in the development of hypoxic brain injury. In this regard, a hallmark feature is microglial activation which results in overproduction of inflammatory cytokines, free radicals and nitric oxide. Concomitantly, activated microglia exhibit enhanced expression of ion channels such as Kv1.2, Kv1.1 and Nav which further promote the release of inflammatory cytokines, chemokines and reactive oxygen species. Through the above-mentioned inflammatory mediators, activated microglia induce neuronal loss, axonal damage and oligodendroglial death along with myelination disturbances. Our recent studies have extended that tumor necrosis factor-alpha, interleukin-1beta, monocyte chemoattractant protein-1 and macrophage colony stimulating factor produced by activated microglia are linked to the pathogenesis of periventricular white matter damage in the hypoxic brain. It is envisaged that a better understanding of the interactions between microglia and neurons, axons and oligodendrocytes is key to the development of effective preventive and therapeutic strategies for mitigation of hypoxic brain injury.


Subject(s)
Brain/growth & development , Hypoxia, Brain/etiology , Hypoxia, Brain/immunology , Hypoxia, Brain/pathology , Microglia/cytology , Microglia/immunology , Axons/pathology , Blood-Brain Barrier/injuries , Brain/immunology , Chemokines/metabolism , Cytokines/metabolism , Humans , Inflammation , Ion Channels/metabolism , Oligodendroglia/pathology , Reactive Oxygen Species/metabolism
15.
Pathol Res Pract ; 207(2): 91-6, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21145174

ABSTRACT

Epitope H contains an O-linked N-acetylglucosamine (O-GlcNAc) residue in a specific conformation and/or environment recognized by mouse IgM monoclonal antibody H (mabH). Epitope H is present in several types of cells and in several polypeptides outside the CNS. Previous results have shown that in the adult human brains, epitope H is confined mostly to a minority of fibrous astrocytes, and it is greatly upregulated in the reactive astrocytes. Post-translational modification with O-GlcNAc occurs on many proteins involved in several cell processes, such as cell cycle progression, apoptosis, proteasome degradation pathways, and modulation of cellular function in response to nutrition and stress. Hypoxia is one of the major causes of cellular stress. Therefore, in this study, we used the mAbH and the indirect immunoperoxidase method to investigate the expression of epitope H in ependymal cells in brains of persons who died with signs of hypoxic encephalopathy. The results of the present study showed that practically all ependymal cells showed cytoplasmic staining for epitope H in supranuclear cytoplasm in the brain of two premature neonates and in ten infants who died with signs of hypoxic encephalopathy. However, the overwhelming majority of ependymal cells of the nine human embryos taken from legal abortions, ranging from 26 days until 13 weeks of gestational age, and of the ten infants' brains without any sign of hypoxic encephalopathy remained negative. Only occasionally did the ependymal cells show weak cytoplasmic staining in some foci. In addition, the reactive astrocytes in the hypoxic brains showed strong cytoplasmic staining, confirming previous results.


Subject(s)
Acetylglucosamine/analysis , Ependyma/immunology , Epitopes/analysis , Fetal Hypoxia/immunology , Hypoxia, Brain/immunology , Antibodies, Monoclonal , Astrocytes/immunology , Cytoplasm/immunology , Ependyma/embryology , Ependyma/pathology , Fetal Hypoxia/pathology , Fluorescent Antibody Technique, Indirect , Gestational Age , Humans , Hypoxia, Brain/embryology , Hypoxia, Brain/mortality , Hypoxia, Brain/pathology , Infant, Newborn , Infant, Premature , Up-Regulation
16.
Gynecol Obstet Fertil ; 38(3): 205-13, 2010 Mar.
Article in French | MEDLINE | ID: mdl-20207183

ABSTRACT

Anti-RhD allo-immunisation has become rare since anti-D prophylaxis was introduced in the seventies; however, it remains the first cause of fetal anemia. It may cause severe fetal complications such as fetal hydrops, cerebral anoxic lesions and fetal death. In the neonatal period, severe jaundices and anemias requiring transfusion or exsanguino-transfusion are still common in case of severe allo-immunisation. Neonatal death and sequellae due to bilirubin encephalopathy have not fully disappeared. Follow-up of pregnancies with maternal allo-immunisation requires identification of the antibody (anti-RhD, anti-Kell and anti-c are the most frequently responsible for fetal complications), dosage and titration. In RhD allo-immunization, feto-maternal incompatibility may be confirmed by non-invasive RHD genotyping of the fetus in maternal blood. In cases at risk for fetal anemia, weekly Doppler assessment of middle cerebral artery peak systolic velocity (MCA-PSV) allows identification of fetal anemia before the occurrence of fetal hydrops. The reference treatment of fetal anemia is in utero fetal transfusion. The risk of fetal loss due to in utero transfusion (IUT) is 3% per procedure. The cumulated risk of fetal loss can thus exceed 10% in case of early occurrence of fetal anemia requiring up to five or six IUTs in a single pregnancy.


Subject(s)
Blood Group Incompatibility , Anemia/embryology , Anemia/therapy , Blood Group Incompatibility/complications , Blood Group Incompatibility/diagnosis , Blood Group Incompatibility/therapy , Blood Transfusion, Intrauterine , Female , Fetal Death/immunology , Fetal Diseases/therapy , Gestational Age , Heart Rate, Fetal , Humans , Hydrops Fetalis/immunology , Hypoxia, Brain/immunology , Infant, Newborn , Kell Blood-Group System/immunology , Kernicterus/immunology , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/embryology , Pregnancy , Prognosis , Rh Isoimmunization/complications , Rh Isoimmunization/prevention & control , Rho(D) Immune Globulin/therapeutic use , Ultrasonography, Prenatal
17.
Free Radic Res ; 44(4): 422-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20166885

ABSTRACT

The objective was to explain the discrepancy in the development of hypoxic ischemic brain injury (HIE) in some asphyxiated newborns rather than others. Forty newborns were classified according to their cerebrospinal neuron-specific-enolase (CSF-NSE) levels on their 5th-day of life; group 1 with low-NSE (n = 25). The remaining 15 newborns had high-NSE and were further divided into a group with no HIE (n = 10, group 2) and another with HIE (n = 5, group 3). CSF-NSE, total-hydroperoxide (TH), biological-antioxidant-potentials (BAPs), 12 cytokines and erythropoietin (EPO) were measured. The TH/BAP gave the oxidative-stress-index (OSI). The BAPs of serial dilutions of three types of EPO were tested. CSF-NSE and TH and mean OSIs were higher in group 3. IL-8 and mean BAPs were higher in group 2 than in group 1. EPO was less detected in group 3. Serial EPO dilutions correlated with their BAPs. Compensatory antioxidants and IL-8 elevation could be protective of perinatal asphyxic brain injury. Antioxidative effect of EPO could be neuroprotective.


Subject(s)
Antioxidants/metabolism , Asphyxia Neonatorum/complications , Hypoxia, Brain/prevention & control , Interleukin-8/cerebrospinal fluid , Oxidative Stress , Asialoglycoproteins/metabolism , Asphyxia Neonatorum/cerebrospinal fluid , Asphyxia Neonatorum/immunology , Erythropoietin/analogs & derivatives , Erythropoietin/cerebrospinal fluid , Erythropoietin/metabolism , Female , Humans , Hydrogen Peroxide/cerebrospinal fluid , Hypoxia, Brain/cerebrospinal fluid , Hypoxia, Brain/etiology , Hypoxia, Brain/immunology , Infant, Newborn , Male , Phosphopyruvate Hydratase/cerebrospinal fluid , Recombinant Proteins , Up-Regulation
18.
Brain Res ; 1259: 80-9, 2009 Mar 09.
Article in English | MEDLINE | ID: mdl-19146835

ABSTRACT

Perinatal hypoxic-ischaemic brain damage is an important cause of neonatal death and permanent neurological impairment. Therapeutic hypothermia may reduce the development of brain damage after hypoxia. Whether to use room-air or 100% oxygen for resuscitation of the asphyxiated neonate is still debated, and there is little knowledge about the combined effects of therapeutic hypothermia and room air resuscitation. We used human NT2-N neurons to test whether oxygen level during reoxygenation would influence the protective effect of hypothermia. Oxygen-glucose deprived (OGD) human NT2-N neurons were exposed to 20 min of low (1%), medium (21%) or high (95%) oxygen concentrations immediately after hypoxia, followed by 20.5 h of hypothermia (33 degrees C) or normothermia (37 degrees C). Cell viability was determined by a methyltetrazolium assay (MTT), cellular energy failure by hypoxanthine release to supernatant, and inflammatory response by the release of IL-8 (Interleukin-8), bFGF (basic fibroblast growth factor), IP-10 (interferon-inducible protein-10) and MCP-1 (monocyte chemotactic protein-1) to supernatant. Post-hypoxic hypothermia resulted in significantly higher MTT cleavage (average 27% of control (SD 11%) vs 24% (SD 12%), p=0.005). Hypoxanthine release was increased both immediately after hypoxia and 21 h later, however less in hypothermic (median increase 2.0 mumol/L, IQR 1.2-3.2) compared to normothermic cells (2.7 mumol/L, IQR 2.1-4.1, p<0.05). All four inflammatory markers increased after hypoxia but not differently between normothermic and hypothermic cells. Oxygen level had no significant effect on cell viability, inflammatory markers or energy status, irrespective of temperature level. We conclude that hypothermia protects isolated neurons after in vitro hypoxia, and that this protection is not affected by hyperoxic, normoxic or hypoxic reoxygenation.


Subject(s)
Cell Survival , Hypothermia, Induced , Hypoxia, Brain/therapy , Neurons/physiology , Oxygen/metabolism , Cell Line , Chemokine CCL2/metabolism , Chemokine CXCL10/metabolism , Fibroblast Growth Factor 2/metabolism , Glucose/deficiency , Humans , Hypoxanthine/metabolism , Hypoxia/immunology , Hypoxia/therapy , Hypoxia, Brain/immunology , Interleukin-8/metabolism , Neurons/immunology , Temperature
19.
Obstet Gynecol ; 112(2 Pt 2): 442-4, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18669757

ABSTRACT

BACKGROUND: The long-term neurological prognosis of severe fetal anemia is usually considered favorable, especially when fetal hydrops regresses after successful in utero transfusion. CASES: We report two cases of prenatally diagnosed fetal cerebral anoxic lesions associated with severe fetal anemia despite appropriate and successful treatment by in utero transfusion. The two pregnancies were terminated. CONCLUSION: Profound fetal anemia may cause anoxic lesions of the fetal brain that may be diagnosed prenatally. If new onset ventriculomegaly is observed on ultrasonography after in utero transfusion for severe fetal anemia, anoxic lesions could be suspected.


Subject(s)
Anemia/immunology , Fetal Diseases/immunology , Hypoxia, Brain/immunology , Kell Blood-Group System/immunology , Rh Isoimmunization/complications , Adult , Anemia/blood , Anemia/diagnostic imaging , Female , Fetal Diseases/blood , Fetal Diseases/diagnostic imaging , Humans , Hypoxia, Brain/blood , Hypoxia, Brain/diagnostic imaging , Pregnancy , Rh Isoimmunization/diagnostic imaging , Ultrasonography, Prenatal
20.
J Neurochem ; 106(5): 2093-105, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18627436

ABSTRACT

Microglial cells are endowed with different potassium ion channels but their expression and specific functions have remained to be fully clarified. This study has shown Kv1.2 expression in the amoeboid microglia in the rat brain between 1 (P1) and 10 (P10) days of age. Kv1.2 expression was localized in the ramified microglia at P14 and was hardly detected at P21. In postnatal rats exposed to hypoxia, Kv1.2 immunoreactivity in microglia was markedly enhanced. Quantitative RT-PCR analysis confirmed Kv1.2 mRNA expression in microglial cells in vitro. It was further shown that Kv1.2 and protein expression coupled with that of interleukin 1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha) was significantly increased when the cells were subjected to hypoxia. The same increase was observed in cells exposed to adenosine 5'-triphosphate (ATP) and lipopolysaccharide (LPS). Concomitantly, the intracellular potassium concentration decreased significantly. Blockade of Kv1.2 channel with rTityustoxin-Kalpha (TsTx) resulted in partial recovery of intracellular potassium concentration accompanied by a reduced expression of IL-1beta and TNF-alpha mRNA and protein expression and intracellular reactive oxygen species (ROS) production. We conclude that Kv1.2 in microglia modulates IL-1beta and TNF-alpha expression and ROS production probably by regulating the intracellular potassium concentration.


Subject(s)
Cytokines/metabolism , Encephalitis/metabolism , Kv1.2 Potassium Channel/physiology , Microglia/metabolism , Oxidative Stress/immunology , Reactive Oxygen Species/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Cell Line , Cell Movement/immunology , Cells, Cultured , Encephalitis/immunology , Encephalitis/physiopathology , Gliosis/immunology , Gliosis/metabolism , Gliosis/physiopathology , Hypoxia, Brain/immunology , Hypoxia, Brain/metabolism , Inflammation Mediators/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Kv1.2 Potassium Channel/drug effects , Kv1.2 Potassium Channel/genetics , Mice , Microglia/immunology , Potassium/metabolism , Potassium Channel Blockers/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...