Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Publication year range
1.
Basic Res Cardiol ; 116(1): 31, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33929610

ABSTRACT

Aircraft noise induces vascular and cerebral inflammation and oxidative stress causing hypertension and cardiovascular/cerebral dysfunction. With the present studies, we sought to determine the role of myeloid cells in the vascular vs. cerebral consequences of exposure to aircraft noise. Toxin-mediated ablation of lysozyme M+ (LysM+) myeloid cells was performed in LysMCreiDTR mice carrying a cre-inducible diphtheria toxin receptor. In the last 4d of toxin treatment, the animals were exposed to noise at maximum and mean sound pressure levels of 85 and 72 dB(A), respectively. Flow cytometry analysis revealed accumulation of CD45+, CD11b+, F4/80+, and Ly6G-Ly6C+ cells in the aortas of noise-exposed mice, which was prevented by LysM+ cell ablation in the periphery, whereas brain infiltrates were even exacerbated upon ablation. Aircraft noise-induced increases in blood pressure and endothelial dysfunction of the aorta and retinal/mesenteric arterioles were almost completely normalized by ablation. Correspondingly, reactive oxygen species in the aorta, heart, and retinal/mesenteric vessels were attenuated in ablated noise-exposed mice, while microglial activation and abundance in the brain was greatly increased. Expression of phagocytic NADPH oxidase (NOX-2) and vascular cell adhesion molecule-1 (VCAM-1) mRNA in the aorta was reduced, while NFκB signaling appeared to be activated in the brain upon ablation. In sum, we show dissociation of cerebral and peripheral inflammatory reactions in response to aircraft noise after LysM+ cell ablation, wherein peripheral myeloid inflammatory cells represent a dominant part of the pathomechanism for noise stress-induced cardiovascular effects and their central nervous counterparts, microglia, as key mediators in stress responses.


Subject(s)
Arteries/enzymology , Brain/enzymology , Encephalitis/prevention & control , Microglia/enzymology , Muramidase/deficiency , Myeloid Cells/enzymology , Noise, Transportation/adverse effects , Peripheral Vascular Diseases/prevention & control , Aircraft , Animals , Arteries/physiopathology , Brain/pathology , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Encephalitis/pathology , Gene Deletion , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Muramidase/genetics , Oxidative Stress , Peripheral Vascular Diseases/enzymology , Peripheral Vascular Diseases/etiology , Peripheral Vascular Diseases/physiopathology , Reactive Oxygen Species/metabolism
2.
J Cell Physiol ; 234(4): 5304-5318, 2019 04.
Article in English | MEDLINE | ID: mdl-30216439

ABSTRACT

Inflammation and apoptosis are two key factors contributing to secondary brain injury after intracerebral hemorrhage (ICH). In the present study, we explored the neuroprotective role of methylene blue (MB) in ICH rats and studied the potential mechanisms involved. Rats were subjected to local injection of collagenase IV in the striatum or sham surgery. We observed that MB treatment could exert a neuroprotective effect on ICH by promoting neurological scores, decreasing the brain water content, alleviating brain-blood barrier disruption, and improving the histological damages in the perihematomal areas. Furthermore, we demonstrated that the various mechanisms underlying MB's neuroprotective effects linked to inhibited apoptosis and inhibited neuroinflammation. In addition, wortmannin, a selective inhibitor of phosphoinositide 3-kinase (PI3K), could reverse the antiapoptotic and anti-inflammatory effects of MB, which suggested that the PI3K-Akt pathway played an important role. In conclusion, these data suggested that MB could inhibit apoptosis and ameliorate neuroinflammation after ICH, and its neuroprotective effects might be exerted via the activation of the PI3K/Akt/GSK3ß pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Cerebral Hemorrhage/drug therapy , Encephalitis/prevention & control , Glycogen Synthase Kinase 3 beta/metabolism , Methylene Blue/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Brain/enzymology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Brain Edema/prevention & control , Capillary Permeability/drug effects , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/enzymology , Cerebral Hemorrhage/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Encephalitis/pathology , Male , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Neurons/enzymology , Neurons/pathology , Neutrophil Infiltration/drug effects , Rats, Sprague-Dawley , Signal Transduction
3.
Radiology ; 290(2): 359-367, 2019 02.
Article in English | MEDLINE | ID: mdl-30615566

ABSTRACT

Purpose To investigate the association of inflammation and brain edema in a cerebral malaria (CM) mouse model with a combination of bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium, referred to as MPO-Gd, and cross-linked iron oxide nanoparticle (CLIO-NP) imaging. Materials and Methods Female wild-type (n = 23) and myeloperoxidase (MPO) knock-out (n = 5) mice were infected with the Plasmodium berghei ANKA strain from May 2016 to July 2018. Seven healthy mice served as control animals. At a Rapid Murine Coma and Behavioral Scale (RMCBS) score of less than 15, mice underwent MRI at 9.4 T and received gadodiamide, MPO-Gd, or CLIO-NPs. T1-weighted MRI was used to assess MPO activity, and T2*-weighted MRI was used to track CLIO-NPs. Immunofluorescent staining and flow cytometric analyses characterized CLIO-NPs, MPO, endothelial cells, and leukocytes. An unpaired, two-tailed Student t test was used to compare groups; Spearman correlation analysis was used to determine the relationship of imaging parameters to clinical severity. Results MPO-Gd enhancement occurred in inflammatory CM hotspots (olfactory bulb > rostral migratory stream > brainstem > cortex, P < .05 for all regions compared with control mice; mean olfactory bulb signal intensity ratio: 1.40 ± 0.07 vs 0.96 ± 0.01, P < .01). The enhancement was reduced in MPO knockout mice (mean signal intensity ratio at 60 minutes: 1.13 ± 0.04 vs 1.40 ± 0.07 in CM, P < .05). Blood-brain barrier compromise was suggested by parenchymal gadolinium enhancement, leukocyte recruitment, and endothelial activation. CLIO-NPs accumulated mainly intravascularly and at the vascular endothelium. CLIO-NPs were also found in the choroid plexus, indicating inflammation of the ventricular system. Blood-cerebrospinal fluid barrier breakdown showed correlation with brain swelling (r2: 0.55, P < .01) and RMCBS score (r2: 0.75, P < .001). Conclusion Iron oxide nanoparticle imaging showed strong inflammatory involvement of the microvasculature in a murine model of cerebral malaria. Furthermore, bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium imaging depicted parenchymal and intraventricular inflammation. This combined molecular imaging approach links vascular inflammation to breakdown of the blood-brain barrier and blood-cerebrospinal fluid barrier that correlate with global brain edema and disease severity. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Kiessling in this issue.


Subject(s)
Brain Edema , Encephalitis , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Malaria, Cerebral , Peroxidase/metabolism , Animals , Brain/diagnostic imaging , Brain/enzymology , Brain/pathology , Brain Edema/diagnostic imaging , Brain Edema/enzymology , Brain Edema/parasitology , Brain Edema/pathology , Disease Models, Animal , Encephalitis/diagnostic imaging , Encephalitis/enzymology , Encephalitis/parasitology , Encephalitis/pathology , Female , Malaria, Cerebral/complications , Malaria, Cerebral/diagnostic imaging , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
J Stroke Cerebrovasc Dis ; 28(12): 104375, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31590996

ABSTRACT

BACKGROUND: Our previous study showed that propofol, one of the widely used anesthetic agents, can attenuate subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) via inhibiting inflammatory and oxidative reaction. However, it is perplexing whether propofol attenuates inflammatory and oxidative reaction through modulating PI3K/Akt pathway. The present study investigated whether PI3K/Akt pathway is involved in propofol's anti-inflammation, antioxidation, and neuroprotection against SAH-induced EBI. MATERIALS AND METHODS: Adult Sprague-Dawley rats underwent SAH and received treatment with propofol or vehicle after 2 and 12 hours of SAH. LY294002 was injected intracerebroventricularly to selectively inhibit PI3K/Akt signaling. Mortality, SAH grading, neurological scores, brain water content, evans blue extravasation, myeloperoxidase, malondialdehyde, superoxide dismutase, and glutathione peroxidase were measured 24 hours after SAH. Immunoreactivity of p-Akt, t-Akt, nuclear factor- kappa B (NF-κB) p65, nuclear factor erythroid-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase (NQO1), and cyclooxygenase-2 (COX-2) in rat brain was determined by western blot. Tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in rat brain were examined by ELISA. RESULTS: Propofol significantly reduces neurological dysfunction, BBB permeability, brain edema, inflammation, and oxidative stress, all of which were reversed by LY294002. Propofol significantly upregulates the immunoreactivity of p-Akt, Nrf2, and NQO1, all of which were abolished by LY294002. Propofol significantly downregulates the overexpression of NF-κB p65, COX-2, TNF-α, and IL-1ß, all of which were inhibited by LY294002. CONCLUSION: These results suggest that propofol attenuates SAH-induced EBI by inhibiting inflammatory reaction and oxidative stress, which might be associated with the activation of PI3K/Akt signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Edema/prevention & control , Brain/drug effects , Encephalitis/prevention & control , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Propofol/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Subarachnoid Hemorrhage/drug therapy , Animals , Brain/enzymology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/pathology , Interleukin-1beta/metabolism , Male , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Pharm Biol ; 57(1): 263-268, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31124385

ABSTRACT

Context: Researchers in a variety of fields have extensively focused on histone deacetylase 6 (HDAC6) due to its aggravation of inflammatory reaction. However, relevant studies examining whether HDAC6 could exacerbate lipopolysaccharide (LPS)-induced inflammation are still lacking. Objective: We assessed the role of HDAC6 in LPS-induced brain inflammation and used the HDAC6-selective inhibitor Tubastatin A (TBSA) to investigate the potential mechanisms further. Materials and methods: Brain inflammation was induced in Kunming (KM) mice via intraperitoneal (I.P.), injection of Lipopolysaccharide (LPS) (1 mg/kg), the TBSA (0.5 mg/kg) was delivered via intraperitoneal. The phosphorylated p38 (p-p38) Mitogen-activated protein kinases (MAPK) and expression of typical inflammatory mediators, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in both the hippocampus and cortex, were examined by immunoblotting. Nissl staining was used to detect the neuronal damage in the hippocampus and the cortex. Results: About 1 mg/kg LPS via daily intraperitoneal (I.P.) injections for 12 days significantly increased p38 MAPK phosphorylation, TNF-α and IL-6 expression, and neuronal loss. However, 0.5 mg/kg TBSA (three days before LPS treatment) by I.P. injections for 15 days could reverse the above results. Conclusions: This present study provided evidence that TBSA significantly suppressed LPS-induced neuroinflammation and the expression of p-p38. Results derived from our study might help reveal the effective targeting strategies of LPS-induced brain inflammation through inhibiting HDAC6.


Subject(s)
Encephalitis/prevention & control , Enzyme Inhibitors/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Lipopolysaccharides , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Encephalitis/enzymology , Inflammation Mediators/metabolism , Male , Mice, Inbred Strains , Phosphorylation
6.
Pak J Pharm Sci ; 32(1(Special)): 391-396, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30852475

ABSTRACT

The aim of this study is to determine the effects of alone or combined usage of doxycycline and meloxicam on brain superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA), and matrix metalloproteinase (MMP)-9 levels of lipopolysaccharide (LPS)-induced brain inflammation. Totally 78 rats were divided into 5 groups; Healthy control (n=6), LPS (n=18, 0.05µg/µL/rat, intracranially), LPS+D (n=18, LPS 0.05µg/µL/rat, intracranially and doxycycline 40 mg/kg, intraperitoneally), LPS+M (n=18, LPS 0.05 µg/µL/rat, intracranially and meloxicam 2 mg/kg, intraperitoneally), LPS+Combination (n=18, LPS 0.05 µg/µL/rat, intracranially and simultaneously both drug combination) groups. Animals were euthanized at 1, 3 and 6 hours following injections and the brains were removed. Brain SOD, CAT, MDA and MMP-9 levels were determined by ELISA reader. Parameters of LPS groups generally different from Healthy control group. When compared to LPS group, increased SOD level of LPS+D at 3 hours and CAT levels of LPS+M and LPS+D groups were determined (P<0.05) at 3 and 6 hours, respectively. In addition, all treatments statistically significantly (P<0.05) decreased MMP-9 levels at 6 hours. In conclusion, doxycycline and meloxicam may show antioxidant effect via increasing antioxidant enzyme production in the brain; however combined usage of drugs may show more beneficial effect for neuroinflammation. .


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antioxidants/metabolism , Cyclooxygenase 2 Inhibitors/therapeutic use , Doxycycline/therapeutic use , Encephalitis/drug therapy , Meloxicam/therapeutic use , Animals , Anti-Bacterial Agents/administration & dosage , Catalase/metabolism , Cyclooxygenase 2 Inhibitors/administration & dosage , Disease Models, Animal , Doxycycline/administration & dosage , Drug Therapy, Combination , Encephalitis/enzymology , Inflammation , Lipopolysaccharides , Male , Meloxicam/administration & dosage , Rats, Wistar , Superoxide Dismutase/metabolism
7.
Biochem Biophys Res Commun ; 503(2): 837-842, 2018 09 05.
Article in English | MEDLINE | ID: mdl-29920240

ABSTRACT

Activation of protein kinase C delta (PKCδ) has been linked to the neuroinflammation but the relationship with the various neurodegenerative diseases including the Alzheimer's disease (AD) was mostly elusive. In the AD brains, the special phospholipids, ethanolamine plasmalogens (Pls), were found to be reduced and our previous study showed that these lipids possess neuroprotective and anti-inflammatory functions. In the present study, we could find that these lipids can significantly attenuate the microglial expression of PKCδ in the neuroinflammation model and in the AD model mice brains. We also show an increase of PKCδ in the human postmortem AD brains. In addition, we also report that scallop derived Pls (sPls) inhibited the p38MAPK and JNK protein expression which are involved in the expressional regulation of PKCδ in the microglial cells. In addition, the lentiviral shRNA-mediated knockdown of PKCδ attenuated the LPS-induced p65 (NF-kB) activation and inflammatory cytokine expression, suggesting that the PKCδ can induce the inflammatory response which can be inhibited by the sPls. Taken together, our recent findings suggest that the sPls can attenuate the increased expression of PKCδ associated with the neuro-inflammation in the murine brain.


Subject(s)
Encephalitis/enzymology , Pectinidae/chemistry , Plasmalogens/pharmacology , Protein Kinase C-delta/metabolism , Animals , Cell Line , Cytokines/metabolism , Encephalitis/genetics , Enzyme Activation/drug effects , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/enzymology , Plasmalogens/administration & dosage , Protein Kinase C-delta/genetics , RNA Interference , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Brain Behav Immun ; 66: 135-145, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28624534

ABSTRACT

Glutaminolysis, a metabolic process that converts glutamine to glutamate, is particularly important for the central nervous system since glutamate is the major transmitter of excitatory synapses. Glutaminase is the mitochondrial enzyme that catalyzes the first step of glutaminolysis. Two genes encode at least four isoforms of glutaminase in humans. Gls1 gene encodes isoforms kidney-type glutaminase (KGA) and glutaminase C (GAC) through alternative splicing, whereas Gls2 gene encodes liver-type glutaminase isoforms. KGA and GAC have been associated with several neurological diseases. However, it remains unclear whether changes in their expressions can directly cause brain abnormalities. Using a transgenic approach, we generated mice that overexpressed GAC in the brain. The resulting transgenic mice had severe impairments in spatial and fear learning compared with littermate controls. The learning deficits were consistent with diminished hippocampal long-term potentiation in the hippocampal slices of the GAC transgenic mice. Furthermore, we found increases in astrocyte and microglia markers, inflammatory factors, and a decrease in synapse marker synaptophysin, suggesting neuroinflammation and synaptic changes in the GAC transgenic mouse brains. In conclusion, these findings provide the first evidence that GAC overexpression in the brain has deleterious effects on learning and synaptic integrity in vivo.


Subject(s)
Brain/enzymology , Conditioning, Classical/physiology , Encephalitis/enzymology , Glutaminase/physiology , Maze Learning/physiology , Synapses/enzymology , Animals , Apoptosis , Encephalitis/etiology , Fear , Glutaminase/metabolism , Hippocampus/enzymology , Hippocampus/physiology , Long-Term Potentiation , Mice , Mice, Transgenic , Neuroglia/enzymology
9.
Biol Pharm Bull ; 40(5): 557-563, 2017.
Article in English | MEDLINE | ID: mdl-28458341

ABSTRACT

Prostaglandin E2 (PGE2) has been thought to be an important mediator of inflammation in peripheral tissues, but recent studies clearly show the involvement of PGE2 in inflammatory brain diseases. In some animal models of brain disease, the genetic disruption and chemical inhibition of cyclooxygenase (COX)-2 resulted in the reduction of PGE2 and amelioration of symptoms, and it had been thought that PGE2 produced by COX-2 may be involved in the progression of injuries. However, COX-2 produces not only PGE2, but also some other prostanoids, and thus the protective effects of COX-2 inhibition, as well as severe side effects, may be caused by the inhibition of prostanoids other than PGE2. Therefore, to elucidate the role of PGE2, studies of microsomal prostaglandin E synthase-1 (mPGES-1), an inducible terminal enzyme for PGE2 synthesis, have recently been an active area of research. Studies from mPGES-1 deficient mice provide compelling evidence for its role in a variety of inflammatory brain diseases, such as ischemic stroke, Alzheimer's disease and epilepsy, and clues for developing new therapeutic treatments for brain diseases by targeting mPGES-1. Considering that COX inhibitors may non-selectively suppress the production of many types of prostanoids that are essential for normal physiological functioning of the brain and peripheral tissues, as well as induce gastro-intestinal, renal and cardiovascular complications, mPGES-1 inhibitors are expected to be injury-selective and have fewer side-effects when treating human brain diseases. Thus, this paper focuses on recent studies that have demonstrated the involvement of mPGES-1 in pathological brain diseases.


Subject(s)
Brain Diseases/genetics , Dinoprostone/metabolism , Encephalitis/genetics , Prostaglandin-E Synthases/genetics , Animals , Brain Diseases/enzymology , Brain Diseases/pathology , Encephalitis/enzymology , Encephalitis/pathology , Humans , Nervous System Diseases/enzymology , Nervous System Diseases/genetics , Nervous System Diseases/pathology
10.
Eur J Neurosci ; 43(1): 41-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26565388

ABSTRACT

Microglia activation and inflammatory factors in brain microenvironment are associated with degeneration of neurons in the substantia nigra (SN) of Parkinson's disease (PD) patients and various PD models. There is increasing evidence that the Rho/ROCK (Rho kinase) signalling pathway may play a critical role in the inflammatory response, and ROCK inhibitor has been reported to have neuroprotective effects. In this study, we examined the neuroprotective potential and possible mechanism of ROCK inhibitor Fasudil in an intranasal lipopolysaccharide (LPS)-induced PD model. ROCK was activated with LPS stimulation and inhibited by Fasudil treatment in this PD model. Behavioural tests demonstrated a clear improvement in motor performance after Fasudil treatment. Furthermore, Fasudil resulted in a significant attenuation of dopamine cell loss, α-synuclein accumulation and inflammatory response with the reversion of inflammatory M1 to anti-inflammatory M2 microglia, decreased NF-кB activation, and IL-12 and TNF-α generation in the SN and olfactory bulb in this model. This study establishes a role for Fasudil in protecting against LPS-mediated dopamine degeneration and provides a therapeutic strategy for the treatment of PD.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Dopaminergic Neurons/drug effects , Encephalitis/enzymology , Encephalitis/prevention & control , Neuroprotective Agents/administration & dosage , Parkinsonian Disorders/complications , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , Administration, Intranasal , Animals , Dopaminergic Neurons/metabolism , Encephalitis/etiology , Female , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/physiology , Motor Activity/drug effects , NF-kappa B/metabolism , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Parkinsonian Disorders/chemically induced , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , alpha-Synuclein/metabolism , rho-Associated Kinases/metabolism
11.
J Neuroinflammation ; 13(1): 86, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27095436

ABSTRACT

BACKGROUND: Protein tyrosine phosphatase 1B (PTP1B) is a member of the non-transmembrane phosphotyrosine phosphatase family. Recently, PTP1B has been proposed to be a novel target of anti-cancer and anti-diabetic drugs. However, the role of PTP1B in the central nervous system is not clearly understood. Therefore, in this study, we sought to define PTP1B's role in brain inflammation. METHODS: PTP1B messenger RNA (mRNA) and protein expression levels were examined in mouse brain and microglial cells after LPS treatment using RT-PCR and western blotting. Pharmacological inhibitors of PTP1B, NF-κB, and Src kinase were used to analyze these signal transduction pathways in microglia. A Griess reaction protocol was used to determine nitric oxide (NO) concentrations in primary microglia cultures and microglial cell lines. Proinflammatory cytokine production was measured by RT-PCR. Western blotting was used to assess Src phosphorylation levels. Immunostaining for Iba-1 was used to determine microglial activation in the mouse brain. RESULTS: PTP1B expression levels were significantly increased in the brain 24 h after LPS injection, suggesting a functional role for PTP1B in brain inflammation. Microglial cells overexpressing PTP1B exhibited an enhanced production of NO and gene expression levels of TNF-α, iNOS, and IL-6 following LPS exposure, suggesting that PTP1B potentiates the microglial proinflammatory response. To confirm the role of PTP1B in neuroinflammation, we employed a highly potent and selective inhibitor of PTP1B (PTP1Bi). In LPS- or TNF-α-stimulated microglial cells, in vitro blockade of PTP1B activity using PTP1Bi markedly attenuated NO production. PTP1Bi also suppressed the expression levels of iNOS, COX-2, TNF-α, and IL-1ß. PTP1B activated Src by dephosphorylating the Src protein at a negative regulatory site. PTP1B-mediated Src activation led to an enhanced proinflammatory response in the microglial cells. An intracerebroventricular injection of PTP1Bi significantly attenuated microglial activation in the hippocampus and cortex of LPS-injected mice compared to vehicle-injected mice. The gene expression levels of proinflammatory cytokines were also significantly suppressed in the brain by a PTP1Bi injection. Together, these data suggest that PTP1Bi has an anti-inflammatory effect in a mouse model of neuroinflammation. CONCLUSIONS: This study demonstrates that PTP1B is an important positive regulator of neuroinflammation and is a promising therapeutic target for neuroinflammatory and neurodegenerative diseases.


Subject(s)
Encephalitis/enzymology , Encephalitis/immunology , Microglia/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microglia/immunology , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 1/immunology , Transfection
12.
Brain Behav Immun ; 51: 109-118, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26254234

ABSTRACT

Microglial activation plays a key role in the development of postoperative cognitive dysfunction (POCD). Nox2, one of the main isoforms of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in the central nervous system, is a predominant source of reactive oxygen species (ROS) overproduction in phagocytes including microglia. We therefore hypothesized that Nox2-induced microglial activation is involved in the development of POCD. Sixteen-month-old C57BL/6 mice were subjected to exploratory laparotomy with isoflurane anesthesia to mimic the clinical human abdominal surgery. Behavioral tests were performed at 6 and 7 d post-surgery with open field and fear conditioning tests, respectively. The levels of Nox2, 8-hydroxy-2'-deoxyguanosine (8-OH-dG, a marker of DNA oxidation), CD11b (a marker of microglial activation), interleukin-1ß (IL-1ß), and brain-derived neurotrophic factor (BDNF) were determined in the hippocampus and prefrontal cortex at 1 d and 7 d post-surgery, respectively. For the interventional study, mice were treated with a NADPH oxidase inhibitor apocynin (APO). Our results showed that exploratory laparotomy with isoflurane anesthesia impaired the contextual fear memory, increased expression of Nox2, 8-OH-dG, CD11b, and IL-1ß, and down-regulated BDNF expression in the hippocampus at 7 d post-surgery. The surgery-induced microglial activation and neuroinflammation persisted to 7 d after surgery in the hippocampus, but only at 1 d in the prefrontal cortex. Notably, administration with APO could rescue these surgery-induced cognitive impairments and associated brain pathology. Together, our data suggested that Nox2-derived ROS in hippocampal microglia, at least in part, contributes to subsequent neuroinflammation and cognitive impairments induced by surgery in aged mice.


Subject(s)
Hippocampus/enzymology , Membrane Glycoproteins/metabolism , Memory Disorders/enzymology , Microglia/enzymology , NADPH Oxidases/metabolism , Postoperative Complications/enzymology , Postoperative Complications/psychology , Reactive Oxygen Species/metabolism , Acetophenones/administration & dosage , Animals , Brain-Derived Neurotrophic Factor/metabolism , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Encephalitis/complications , Encephalitis/enzymology , Enzyme Inhibitors/administration & dosage , Fear/drug effects , Fear/physiology , Hippocampus/drug effects , Laparotomy , Male , Membrane Glycoproteins/antagonists & inhibitors , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Motor Activity/drug effects , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology
13.
J Biol Regul Homeost Agents ; 29(1): 245-50, 2015.
Article in English | MEDLINE | ID: mdl-25864766

ABSTRACT

Dolphin Morbillivirus (DMV), Toxoplasma gondii and Brucella ceti are pathogens of major concern for wild cetaceans. Although a more or less severe encephalitis/meningo-encephalitis may occur in striped dolphins (Stenella coeruleoalba) and bottlenose dolphins (Tursiops truncatus) infected by the aforementioned agents, almost no information is available on the neuropathogenesis of brain lesions, including the neuronal and non-neuronal cells targeted during infection, along with the mechanisms underlying neurodegeneration. We analyzed 5-lipoxygenase (5-LOX) expression in the brain of 11 striped dolphins and 5 bottlenose dolphins, affected or not by encephalitic lesions of various degrees associated with DMV, T. gondii and B. ceti. All the 8 striped dolphins with encephalitis showed a more consistent 5-LOX expression than that observed in the 3 striped dolphins showing no morphologic evidence of brain lesions, with the most prominent band intensity being detected in a B. ceti-infected animal. Similar results were not obtained in T. gondii-infected vs T. gondii-uninfected bottlenose dolphins. Overall, the higher 5-LOX expression found in the brain of the 8 striped dolphins with infectious neuroinflammation is of interest, given that 5-LOX is a putative marker for neurodegeneration in human patients and in experimental animal models. Therefore, further investigation on this challenging issue is also needed in stranded cetaceans affected by central neuropathies.


Subject(s)
Arachidonate 5-Lipoxygenase/analysis , Bottle-Nosed Dolphin , Brain/enzymology , Brain/pathology , Encephalitis/veterinary , Stenella , Animals , Blotting, Western , Brain/microbiology , Brain/virology , Brucella/pathogenicity , Brucellosis/microbiology , Brucellosis/pathology , Brucellosis/veterinary , Encephalitis/enzymology , Encephalitis/virology , Meningoencephalitis/enzymology , Meningoencephalitis/pathology , Meningoencephalitis/veterinary , Morbillivirus/pathogenicity , Morbillivirus Infections/veterinary , Morbillivirus Infections/virology , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/enzymology , Toxoplasmosis, Animal/pathology
14.
Ann Neurol ; 73(1): 120-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23225603

ABSTRACT

OBJECTIVE: To report a novel cell surface autoantigen of encephalitis that is a critical regulatory subunit of the Kv4.2 potassium channels. METHODS: Four patients with encephalitis of unclear etiology and antibodies with a similar pattern of neuropil brain immunostaining were selected for autoantigen characterization. Techniques included immunoprecipitation, mass spectrometry, cell-base experiments with Kv4.2 and several dipeptidyl-peptidase-like protein-6 (DPPX) plasmid constructs, and comparative brain immunostaining of wild-type and DPPX-null mice. RESULTS: Immunoprecipitation studies identified DPPX as the target autoantigen. A cell-based assay confirmed that all 4 patients, but not 210 controls, had DPPX antibodies. Symptoms included agitation, confusion, myoclonus, tremor, and seizures (1 case with prominent startle response). All patients had pleocytosis, and 3 had severe prodromal diarrhea of unknown etiology. Given that DPPX tunes up the Kv4.2 potassium channels (involved in somatodendritic signal integration and attenuation of dendritic back-propagation of action potentials), we determined the epitope distribution in DPPX, DPP10 (a protein homologous to DPPX), and Kv4.2. Patients' antibodies were found to be specific for DPPX, without reacting with DPP10 or Kv4.2. The unexplained diarrhea led to a demonstration of a robust expression of DPPX in the myenteric plexus, which strongly reacted with patients' antibodies. The course of neuropsychiatric symptoms was prolonged and often associated with relapses during decreasing immunotherapy. Long-term follow-up showed substantial improvement in 3 patients (1 was lost to follow-up). INTERPRETATION: Antibodies to DPPX are associated with a protracted encephalitis characterized by central nervous system hyperexcitability (agitation, myoclonus, tremor, seizures), pleocytosis, and frequent diarrhea at symptom onset. The disorder is potentially treatable with immunotherapy.


Subject(s)
Autoantibodies/biosynthesis , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/immunology , Encephalitis/immunology , Nerve Tissue Proteins/immunology , Potassium Channels/immunology , Shal Potassium Channels/metabolism , Aged , Animals , Antigen-Antibody Reactions/immunology , Autoantibodies/chemistry , Encephalitis/enzymology , Encephalitis/pathology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , Shal Potassium Channels/chemistry , Shal Potassium Channels/immunology
15.
Toxicol Appl Pharmacol ; 281(2): 185-94, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25448683

ABSTRACT

Tetramethylenedisulfotetramine (TETS) is a potent convulsant poison for which there is currently no approved antidote. The convulsant action of TETS is thought to be mediated by inhibition of type A gamma-aminobutyric acid receptor (GABAAR) function. We, therefore, investigated the effects of post-exposure administration of diazepam, a GABAAR positive allosteric modulator, on seizure activity, death and neuroinflammation in adult male Swiss mice injected with a lethal dose of TETS (0.15mg/kg, ip). Administration of a high dose of diazepam (5mg/kg, ip) immediately following the second clonic seizure (approximately 20min post-TETS injection) effectively prevented progression to tonic seizures and death. However, this treatment did not prevent persistent reactive astrogliosis and microglial activation, as determined by GFAP and Iba-1 immunoreactivity and microglial cell morphology. Inhibition of soluble epoxide hydrolase (sEH) has been shown to exert potent anti-inflammatory effects and to increase survival in mice intoxicated with other GABAAR antagonists. The sEH inhibitor TUPS (1mg/kg, ip) administered immediately after the second clonic seizure did not protect TETS-intoxicated animals from tonic seizures or death. Combined administration of diazepam (5mg/kg, ip) and TUPS (1mg/kg, ip, starting 1h after diazepam and repeated every 24h) prevented TETS-induced lethality and influenced signs of neuroinflammation in some brain regions. Significantly decreased microglial activation and enhanced reactive astrogliosis were observed in the hippocampus, with no changes in the cortex. Combining an agent that targets specific anti-inflammatory mechanisms with a traditional antiseizure drug may enhance treatment outcome in TETS intoxication.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anticonvulsants/administration & dosage , Brain/drug effects , Bridged-Ring Compounds , Diazepam/administration & dosage , Encephalitis/prevention & control , Enzyme Inhibitors/administration & dosage , Epoxide Hydrolases/antagonists & inhibitors , GABA Modulators/administration & dosage , Phenylurea Compounds/administration & dosage , Piperidines/administration & dosage , Seizures/prevention & control , Animals , Brain/enzymology , Brain/physiopathology , Brain Waves/drug effects , Disease Models, Animal , Drug Administration Schedule , Drug Therapy, Combination , Electroencephalography , Encephalitis/chemically induced , Encephalitis/enzymology , Encephalitis/physiopathology , Epoxide Hydrolases/metabolism , Male , Mice , Seizures/chemically induced , Seizures/enzymology , Seizures/physiopathology , Time Factors
16.
BMC Neurosci ; 14: 26, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23497276

ABSTRACT

BACKGROUND: Chronic administration of Aluminum is proposed as an environmental factor that may affect several enzymes and other biomolecules related to neurotoxicity and Alzheimer's disease (AD). APE1 a multifunctional protein, functions in DNA repair and plays a key role in cell survival versus cell death upon stimulation with cytotoxic agent, making it an attractive emerging therapeutic target. The promising protective effect of resveratrol (resv), which is known to exert potent anti-inflammatory effects on neurotoxicity induced by aluminum chloride (AlCl3), may be derived from its own antioxidant properties. In the present work we investigated the modulation of APE1 expression during AlCl3-induced neuroinflammation (25 mg/Kg body weight by oral gavages) in experimental rats. We tested the hypothesis that a reactive oxygen species (ROS)-scavenger, resveratrol at 0.5 mg/kg bodyweight, which is known to exert potent anti-inflammatory effects, would attenuate central inflammation and modulate APE1 expression in AlCl3-fed rats. Neuroinflammation-induced genes including ß-secretase (BACE), amyloid-ß precursor protein (APP), presenilin 2 (PSEN-2) and sirt-2 were determined by RT-PCR. APE1 is determined at mRNA and protein levels and confirmed by immunohistochemistry. The expression of pro-inflammatory cytokines (TNF-α, IL6) and iNOS by the rat brain extract were measured by RT-PCR. RESULT: Our results indicate that resveratrol may attenuate AlCl3-induced direct neuroinflammation in rats, and its mechanisms are, at least partly, due to maintaining high APE1 level. Resveratrol co-administration with aluminum chloride exerted more protective effect than pre-administration or treatment of induced rats. A significant elevation of APE1 at both mRNA and protein levels was observed in addition to a marked reduction in ß-secretase and amyloid-ß. We found that AlCl3 stimulated the expression of TNF-α, IL-6, and iNOS in rat brain in which NF-κB was involved. Resveratrol inhibited AlCl3-induced expression and release of TNF-α, IL-6, and iNOS in rat brain. CONCLUSIONS: These findings establish a role for APE1 as a master regulator of AlCl3 dependent inflammatory responses in rat brain. In addition, there was an ameliorative change with resveratrol against AlCl3-induced neurotoxicity. These results suggest that rat brain cells produce pro-inflammatory cytokines in response to AlCl3 in a similar pattern, and further suggest that resveratrol exerts anti-inflammatory effects in rat brain, at least partly, by inhibiting different pro-inflammatory cytokines and key signaling molecules. It might be a potential agent for treatment of neuroinflammation-related diseases, such as AD.


Subject(s)
Aluminum Compounds/toxicity , Chlorides/toxicity , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Encephalitis/chemically induced , Encephalitis/enzymology , Aluminum Chloride , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartate Aminotransferases/metabolism , Catalase/metabolism , Disease Models, Animal , Glutathione/metabolism , Glutathione Transferase/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipid Peroxidation/drug effects , Male , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Peptide Fragments/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Rats , Rats, Wistar
17.
J Neurosci ; 31(42): 14850-60, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22016518

ABSTRACT

Cyclooxygenase-2 (COX-2), a source of inflammatory mediators and a multifunctional neuronal modulator, is rapidly induced in select populations of cortical neurons after status epilepticus. The consequences of rapid activity-triggered induction of COX-2 in neurons have been the subject of much study and speculation. To address this issue directly, we created a mouse in which COX-2 is conditionally ablated in selected forebrain neurons. Results following pilocarpine-induced status epilepticus indicate that neuronal COX-2 promotes early neuroprotection and then delayed neurodegeneration of CA1 pyramidal neurons, promotes neurodegeneration of nearby somatostatin interneurons in the CA1 stratum oriens and dentate hilus (which themselves do not express COX-2), intensifies a broad inflammatory reaction involving numerous cytokines and other inflammatory mediators in the hippocampus, and is essential for development of a leaky blood-brain barrier after seizures. These findings point to a profound role of seizure-induced neuronal COX-2 expression in neuropathologies that accompany epileptogenesis.


Subject(s)
Cyclooxygenase 2/deficiency , Encephalitis/enzymology , Encephalitis/prevention & control , Neurons/pathology , Prosencephalon/pathology , Status Epilepticus/complications , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Blood-Testis Barrier/parasitology , Cyclooxygenase 2/genetics , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Electroencephalography/methods , Electromyography/methods , Encephalitis/etiology , Encephalitis/pathology , Fluoresceins , Functional Laterality , Gene Expression Regulation/genetics , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Agonists/toxicity , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Organic Chemicals , Pilocarpine/toxicity , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Somatostatin/metabolism , Status Epilepticus/chemically induced
18.
J Neuroinflammation ; 9: 82, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22540228

ABSTRACT

BACKGROUND: The mechanisms of progressive dopaminergic neuronal loss in Parkinson's disease (PD) remain poorly understood, largely due to the complex etiology and multifactorial nature of disease pathogenesis. Several lines of evidence from human studies and experimental models over the last decade have identified neuroinflammation as a potential pathophysiological mechanism contributing to disease progression. Tumor necrosis factor α (TNF) has recently emerged as the primary neuroinflammatory mediator that can elicit dopaminergic cell death in PD. However, the signaling pathways by which TNF mediates dopaminergic cell death have not been completely elucidated. METHODS: In this study we used a dopaminergic neuronal cell model and recombinant TNF to characterize intracellular signaling pathways activated during TNF-induced dopaminergic neurotoxicity. Etanercept and neutralizing antibodies to tumor necrosis factor receptor 1 (TNFR1) were used to block TNF signaling. We confirmed the results from our mechanistic studies in primary embryonic mesencephalic cultures and in vivo using the stereotaxic lipopolysaccharide (LPS) model of nigral dopaminergic degeneration. RESULTS: TNF signaling in dopaminergic neuronal cells triggered the activation of protein kinase Cδ (PKCδ), an isoform of the novel PKC family, by caspase-3 and caspase-8 dependent proteolytic cleavage. Both TNFR1 neutralizing antibodies and the soluble TNF receptor Etanercept blocked TNF-induced PKCδ proteolytic activation. Proteolytic activation of PKCδ was accompanied by translocation of the kinase to the nucleus. Notably, inhibition of PKCδ signaling by small interfering (si)RNA or overexpression of a PKCδ cleavage-resistant mutant protected against TNF-induced dopaminergic neuronal cell death. Further, primary dopaminergic neurons obtained from PKCδ knockout (-/-) mice were resistant to TNF toxicity. The proteolytic activation of PKCδ in the mouse substantia nigra in the neuroinflammatory LPS model was also observed. CONCLUSIONS: Collectively, these results identify proteolytic activation of PKCδ proapoptotic signaling as a key downstream effector of dopaminergic cell death induced by TNF. These findings also provide a rationale for therapeutically targeting PKCδ to mitigate progressive dopaminergic degeneration resulting from chronic neuroinflammatory processes.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Dopaminergic Neurons/enzymology , Encephalitis/enzymology , Encephalitis/pathology , Protein Kinase C-delta/metabolism , Receptors, Death Domain/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/toxicity , Cell Death/physiology , Cells, Cultured , Dopaminergic Neurons/pathology , Encephalitis/etiology , Enzyme Activation/physiology , Mice , Protein Kinase C-delta/physiology , Proteolysis , Rats , Tumor Necrosis Factor-alpha/toxicity
19.
Neurobiol Dis ; 42(3): 341-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21303700

ABSTRACT

Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is widely expressed in brain tissue including neurons, glia, and endothelia in neurovascular units. It is a major source of oxidants in the post-ischemic brain and significantly contributes to ischemic brain damage. Inflammation occurs after brain ischemia and is known to be associated with post-ischemic oxidative stress. Post-ischemic inflammation also causes progressive brain injury. In this study we investigated the role of NOX2 in post-ischemic cerebral inflammation using a transient middle cerebral artery occlusion model in mice. We demonstrate that mice with NOX2 subunit gp91(phox) knockout (gp91 KO) showed 35-44% less brain infarction at 1 and 3 days of reperfusion compared with wild-type (WT) mice. Minocycline further reduced brain damage in the gp91 KO mice at 3 days of reperfusion. The gp91 KO mice exhibited less severe post-ischemic inflammation in the brain, as evidenced by reduced microglial activation and decreased upregulation of inflammation mediators, including interleukin-1ß (IL-1ß), tumor necrosis factor-α, inducible nitric oxide synthases, CC-chemokine ligand 2, and CC-chemokine ligand 3. Finally, we demonstrated that an intraventricular injection of IL-1ß enhanced ischemia- and reperfusion-mediated brain damage in the WT mice (double the infarction volume), whereas, it failed to aggravate brain infarction in the gp91 KO mice. Taken together, these results demonstrate the involvement of NOX2 in post-ischemic neuroinflammation and that NOX2 inhibition provides neuroprotection against inflammatory cytokine-mediated brain damage.


Subject(s)
Brain Ischemia/enzymology , Brain/enzymology , Encephalitis/enzymology , NADPH Oxidases/metabolism , Analysis of Variance , Animals , Blotting, Western , Brain/drug effects , Brain Ischemia/complications , Brain Ischemia/drug therapy , Cytokines/metabolism , Encephalitis/drug therapy , Encephalitis/etiology , Immunohistochemistry , Mice , Mice, Knockout , Minocycline/pharmacology , Minocycline/therapeutic use , Oxidative Stress/physiology , Reverse Transcriptase Polymerase Chain Reaction
20.
J Neuroinflammation ; 8: 81, 2011 Jul 18.
Article in English | MEDLINE | ID: mdl-21767382

ABSTRACT

BACKGROUND: Estrogens from peripheral sources as well as central aromatization are neuroprotective in the vertebrate brain. Under normal conditions, aromatase is only expressed in neurons, however following anoxic/ischemic or mechanical brain injury; aromatase is also found in astroglia. This increased glial aromatization and the consequent estrogen synthesis is neuroprotective and may promote neuronal survival and repair. While the effects of estradiol on neuroprotection are well studied, what induces glial aromatase expression remains unknown. METHODS: Adult male zebra finches (Taeniopygia guttata) were given a penetrating injury to the entopallium. At several timepoints later, expression of aromatase, IL-1ß-like, and IL-6-like were examined using immunohistochemistry. A second set of zebra birds were exposed to phytohemagglutinin (PHA), an inflammatory agent, directly on the dorsal surface of the telencephalon without creating a penetrating injury. Expression of aromatase, IL-1ß-like, and IL-6-like were examined using both quantitative real-time polymerase chain reaction to examine mRNA expression and immunohistochemistry to determine cellular expression. Statistical significance was determined using t-test or one-way analysis of variance followed by the Tukey Kramers post hoc test. RESULTS: Following injury in the zebra finch brain, cytokine expression occurs prior to aromatase expression. This temporal pattern suggests that cytokines may induce aromatase expression in the damaged zebra finch brain. Furthermore, evoking a neuroinflammatory response characterized by an increase in cytokine expression in the uninjured brain is sufficient to induce glial aromatase expression. CONCLUSIONS: These studies are among the first to examine a neuroinflammatory response in the songbird brain following mechanical brain injury and to describe a novel neuroimmune signal to initiate aromatase expression in glia.


Subject(s)
Aromatase/metabolism , Brain/cytology , Brain/enzymology , Encephalitis/enzymology , Encephalitis/pathology , Finches , Neuroglia/enzymology , Animals , Aromatase/genetics , Biomarkers/metabolism , Brain/anatomy & histology , Brain/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Cell Death/physiology , Encephalitis/chemically induced , Estrogens/metabolism , Finches/anatomy & histology , Finches/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Neuroglia/cytology , Neuroprotective Agents/metabolism , Phytohemagglutinins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL