Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 193
1.
Basic Res Cardiol ; 116(1): 31, 2021 04 30.
Article En | MEDLINE | ID: mdl-33929610

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.


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.
Neurotoxicology ; 83: 77-88, 2021 03.
Article En | MEDLINE | ID: mdl-33417987

Low-dose repeated lipopolysaccharide pre-challenge followed by chronic mild stress (LPS/CMS) protocol has been introduced as a rodent model of depression combining the roles of immune activation and chronic psychological stress. However, the impact of this paradigm on cognitive functioning has not been investigated hitherto. METHODS: This study evaluated LPS/CMS-induced cognitive effects and the role of glycogen synthase kinase-3ß (GSK-3ß) activation with subsequent neuroinflammation and pathological tau deposition in the pathogenesis of these effects using lithium (Li) as a tool for GSK-3 inhibition. RESULTS: LPS pre-challenge reduced CMS-induced neuroinflammation, depressive-like behavior and cognitive inflexibility. It also improved spatial learning but increased GSK-3ß expression and exaggerated hyperphosphorylated tau accumulation in hippocampus and prefrontal cortex. Li ameliorated CMS and LPS/CMS-induced depressive and cognitive deficits, reduced GSK-3ß over-expression and tau hyperphosphorylation, impeded neuroinflammation and enhanced neuronal survival. CONCLUSION: This study draws attention to LPS/CMS-triggered cognitive changes and highlights how prior low-dose immune challenge could develop an adaptive capacity to buffer inflammatory damage and maintain the cognitive abilities necessary to withstand threats. This work also underscores the favorable effect of Li (as a GSK-3ß inhibitor) in impeding exaggerated tauopathy and neuroinflammation, rescuing neuronal survival and preserving cognitive functions. Yet, further in-depth studies utilizing different low-dose LPS challenge schedules are needed to elucidate the complex interactions between immune activation and chronic stress exposure.


Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Depression/prevention & control , Encephalitis/prevention & control , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hippocampus/drug effects , Lithium Chloride/pharmacology , Protein Kinase Inhibitors/pharmacology , Tauopathies/prevention & control , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/physiopathology , Chronic Disease , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Depression/enzymology , Depression/etiology , Depression/physiopathology , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Encephalitis/physiopathology , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/enzymology , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Phosphorylation , Rats, Wistar , Spatial Learning/drug effects , Stress, Psychological/complications , Stress, Psychological/psychology , Tauopathies/enzymology , Tauopathies/etiology , Tauopathies/physiopathology , tau Proteins/metabolism
3.
Toxicol Lett ; 318: 44-49, 2020 Jan.
Article En | MEDLINE | ID: mdl-31639409

Acrolein is a neurotoxin produced through lipid peroxidation in the brain affected by ischemic stroke, which results in neuronal cell injury and inflammation. However the mechanism underlying acrolein-induced brain inflammation remains unclear. Therefore we examined how acrolein leads to astrocytic inflammation. It was found that acrolein increased the levels of NLRP3 and cleaved caspase-1, which led to the maturation of interleukin-1ß (IL-1ß). ELISA assay results, which showed that acrolein increased the secreted IL-1ß, further supported acrolein-induced astrocytic inflammation. Acrolein increased ADAM10 protein levels and the cleavage of N-cadherin. The ADAM10 inhibitor, GI 254023X blocked N-cadherin cleavage by acrolein, suggesting that ADAM10 is an upstream of N-cadherin. Furthermore, we found that acrolein activated p38 MAPK and NF-κB p65, while pretreatment with p38 MAPK inhibitor, SB203580 and GI 254023X inhibited NF-κB p65 activation and NLRP3 inflammasome. This suggests that p38 MAPK mediates the activation of NF-κB p65, which is associated with NLRP3 expression. Finally, we showed that acrolein induced cell toxicity and decrease of EAAT1 expression, suggesting that acrolein may induce a loss of glutamate uptake function. In conclusion, we demonstrate that acrolein induces astrocytic inflammation through NLRP3 inflammasome, which is regulated by ADAM10 and attributed to p38 MAPK-activated NF-κB p65 activity.


ADAM10 Protein/metabolism , Acrolein/toxicity , Apoptosis/drug effects , Astrocytes/drug effects , Brain/drug effects , Encephalitis/chemically induced , Inflammasomes/drug effects , ADAM10 Protein/genetics , Animals , Astrocytes/enzymology , Astrocytes/pathology , Brain/enzymology , Brain/pathology , Cadherins/metabolism , Caspase 1/metabolism , Cell Line , Encephalitis/enzymology , Encephalitis/pathology , Excitatory Amino Acid Transporter 1/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nerve Tissue Proteins/metabolism , Rats , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Neurochem Int ; 131: 104563, 2019 12.
Article En | MEDLINE | ID: mdl-31589911

Peripheral lipopolysaccharide (LPS) injection induces systemic inflammation through the activation of the inhibitor of nuclear factor kappa B (NF-κB) kinase (IKK)/NF-κB signaling pathway, which promotes brain dysfunction resulting in conditions including anorexia. LPS-mediated reduction of food intake is associated with activation of NF-κB signaling and phosphorylation of the transcription factor signal transducer and activator of transcription 3 (STAT3) in the hypothalamus. We recently reported phospholipase C-related catalytically inactive protein (PRIP) as a new negative regulator of phosphatidylinositol 3-kinase/AKT signaling. AKT regulates the IKK/NF-κB signaling pathway; therefore, this study aimed to investigate the role of PRIP/AKT signaling in LPS-mediated neuroinflammation-induced anorexia. PRIP gene (Prip1 and Prip2) knockout (Prip-KO) mice intraperitoneally (ip) administered with LPS exhibited increased anorexia responses compared with wild-type (WT) controls. Although few differences were observed between WT and Prip-KO mice in LPS-elicited plasma pro-inflammatory cytokine elevation, hypothalamic pro-inflammatory cytokines were significantly upregulated in Prip-KO rather than WT mice. Hypothalamic AKT and IKK phosphorylation and IκB degradation were significantly increased in Prip-KO rather than WT mice, indicating further promotion of AKT-mediated NF-κB signaling. Consistently, hypothalamic STAT3 was further phosphorylated in Prip-KO rather than WT mice. Furthermore, suppressor of cytokine signaling 3 (Socs3), a negative feedback regulator for STAT3 signaling, and cyclooxogenase-2 (Cox2), a candidate molecule in LPS-induced anorexigenic responses, were upregulated in the hypothalamus in Prip-KO rather than WT mice. Pro-inflammatory cytokines were upregulated in hypothalamic microglia isolated from Prip-KO rather than WT mice. Together, these findings indicate that PRIP negatively regulates LPS-induced anorexia caused by pro-inflammatory cytokine expression in the hypothalamus, which is mediated by AKT-activated NF-κB signaling. Importantly, hypothalamic microglia participate in this PRIP-mediated process. Elucidation of PRIP-mediated neuroinflammatory responses may provide novel insights into the pathophysiology of many brain dysfunctions.


Anorexia/enzymology , Encephalitis/enzymology , Hypothalamus/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Anorexia/chemically induced , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Eating , Encephalitis/chemically induced , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
5.
J Stroke Cerebrovasc Dis ; 28(12): 104375, 2019 Dec.
Article En | MEDLINE | ID: mdl-31590996

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.


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
6.
Pharm Biol ; 57(1): 263-268, 2019 Dec.
Article En | MEDLINE | ID: mdl-31124385

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.


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
7.
Mil Med ; 184(Suppl 1): 265-272, 2019 03 01.
Article En | MEDLINE | ID: mdl-30901455

OBJECTIVE: Mild blast traumatic brain injury is commonly prevalent in modern combat casualty care and has been associated with the development of neurodegenerative conditions. However, whether primary lower level blast overpressure (LBOP) causes neurodegeneration and neuroinflammation remains largely unknown. The aim of our present study was to determine whether LBOP can cause neuroinflammation and neurodegeneration. METHODS: Anesthetized rats were randomly assigned to LBOP group (70 kPa, n = 5) or sham group (without blast, n = 5). Histopathological and cytokine changes in brain tissue at 5 days post-injury were evaluated by hematoxylin-eosin staining and Bioplex assay, respectively. RESULTS: Histopathological assessment revealed neuronal degeneration and increased density of inflammatory cells in frontal and parietal cortex, hippocampus and thalamus in rats exposed to LBOP. LBOP exposure significantly elevated levels of pro-inflammatory cytokines (EPO, IL-1ß, IL-6, IL-12, IL-18, and TNF-α) and chemokines (GRO and RANTES) as well as of an anti-inflammatory cytokine (IL-13) in the frontal cortex. CONCLUSIONS: This study reveals a role of neuroinflammation in neurodegeneration after mild blast traumatic brain injury. Therapies that target this process might in warfighters might function either by attenuating the development of post-traumatic stress disorder, chronic traumatic encephalopathy and Alzheimer's disease, or by slowing their progression.


Encephalitis/pathology , Explosions/statistics & numerical data , Nerve Degeneration/pathology , Animals , Biomarkers/analysis , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Chemokine CCL5/analysis , Chemokine CXCL1/analysis , Chemokines/analysis , Cytokines/analysis , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Interleukin-12/analysis , Interleukin-18/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Rats/injuries , Tumor Necrosis Factor-alpha/analysis
8.
Pak J Pharm Sci ; 32(1(Special)): 391-396, 2019 Jan.
Article En | MEDLINE | ID: mdl-30852475

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. .


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
9.
Radiology ; 290(2): 359-367, 2019 02.
Article En | MEDLINE | ID: mdl-30615566

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.


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
10.
J Cell Physiol ; 234(4): 5304-5318, 2019 04.
Article En | MEDLINE | ID: mdl-30216439

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.


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
11.
Toxicol Sci ; 168(2): 381-393, 2019 04 01.
Article En | MEDLINE | ID: mdl-30576531

Spermine oxidase (Smox) is a member of the polyamine oxidases and has been demonstrated to be involved in ischemic brain damage. In this study, we found that Smox expression was increased in a rat middle cerebral artery occlusion (MCAO) model and in cultured primary neurons after oxygen-glucose deprivation and reoxygenation (OGD/R). Smox downregulation by the adeno-associated virus RNA interference system significantly reduced the MCAO-induced brain infarct volume and neurological deficits and decreased neuronal apoptosis and inflammatory reactions. In addition, significant microglial activation and increased IL-6 and TNF-α expression were observed in microglia treated with supernatant from neurons after OGD/R. However, a significant reduction in microglial activation as well as IL-6 and TNF-α expression was observed in microglia treated with supernatant from Smox downregulated neurons after OGD/R. Therefore, the results indicated that Smox is an important mediator of cerebral ischemia injury and may be a therapeutic target for cerebral ischemia patients.


Brain Ischemia/prevention & control , Encephalitis/prevention & control , Oxidoreductases Acting on CH-NH Group Donors/genetics , Reperfusion Injury/prevention & control , Animals , Apoptosis/genetics , Brain Ischemia/enzymology , Cells, Cultured , Disease Models, Animal , Down-Regulation , Encephalitis/enzymology , Infarction, Middle Cerebral Artery , Interleukin-6/genetics , Male , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Tumor Necrosis Factor-alpha/genetics , Polyamine Oxidase
12.
Biochem Biophys Res Commun ; 503(2): 837-842, 2018 09 05.
Article En | MEDLINE | ID: mdl-29920240

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.


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
13.
Appl Radiat Isot ; 132: 6-12, 2018 Feb.
Article En | MEDLINE | ID: mdl-29127936

The reference standard N-(3-(4-methylpiperazin-1-yl)-1-(5-methylpyridin-2-yl)-1H-pyrazol-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (9) and its demethylated precursor N-(1-(5-methylpyridin-2-yl)-3-(piperazin-1-yl)-1H-pyrazol-5-yl)pyrazolo[1,5-α]pyrimidine-3-carboxamide (8) were synthesized from pyrazolo[1,5-a]pyrimidine-3-carboxylic acid and ethyl 2-cyanoacetate with overall chemical yield 13% in nine steps and 14% in eight steps, respectively. The target tracer N-(3-(4-[11C]methylpiperazin-1-yl)-1-(5-methylpyridin-2-yl)-1H-pyrazol-5-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide ([11C]9) was prepared from its precursor with [11C]CH3OTf through N-[11C]methylation and isolated by HPLC combined with SPE in 50-60% radiochemical yield, based on [11C]CO2 and decay corrected to EOB. The radiochemical purity was >99%, and the specific activity at EOB was 370-1110 GBq/µmol.


Carbon Radioisotopes/chemistry , Contrast Media/chemical synthesis , Encephalitis/diagnostic imaging , Interleukin-1 Receptor-Associated Kinases/metabolism , Positron-Emission Tomography , Pyrazoles/chemical synthesis , Pyrimidines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Encephalitis/enzymology , Humans , Solid Phase Extraction , Spectrometry, Mass, Electrospray Ionization
14.
Toxicol Sci ; 160(1): 4-14, 2017 Nov 01.
Article En | MEDLINE | ID: mdl-29036520

The lung is constantly exposed to ambient pollutants such as ambient fine particulate matter (PM2.5), making it one of the most frequent locations of inflammation in the body. Given the establishment of crucial role of inflammation in the pathogenesis of cardiometabolic diseases, pulmonary inflammation is thus widely believed to be an important risk factor for cardiometabolic diseases. However, the causality between them has not yet been well established. To determine if pulmonary inflammation is sufficient to cause adverse cardiometabolic effects, SFTPC-rtTA+/-tetO-cre+/-pROSA-inhibitor κB kinase 2(IKK2)ca+/- (LungIKK2ca) and littermate SFTPC-rtTA+/-tetO-cre-/-pROSA-IKK2ca+/- wildtype (WT) mice were fed with doxycycline diet to induce constitutively active Ikk2 (Ikk2ca) overexpression in the lung and their pulmonary, systemic, adipose, and hypothalamic inflammations, vascular function, and glucose homeostasis were assessed. Feeding with doxycycline diet resulted in IKK2ca overexpression in the lungs of LungIKK2ca but not WT mice. This induction of IKK2ca was accompanied by marked pulmonary inflammation as evidenced by significant increases in bronchoalveolar lavage fluid leukocytes, pulmonary macrophage infiltration, and pulmonary mRNA expression of tumor necrosis factor α (Tnfα) and interleukin-6 (Il-6). This pulmonary inflammation due to lung-specific overexpression of IKK2ca was sufficient to increase circulating TNFα and IL-6 levels, adipose expression of Tnfα and Il-6 mRNA, aortic endothelial dysfunction, and systemic insulin resistance. Unexpectedly, no significant alteration in hypothalamic expression of Tnfα and Il-6 mRNA and glucose intolerance were observed in these mice. Pulmonary inflammation is sufficient to induce systemic inflammation, endothelial dysfunction, and insulin resistance, but not hypothalamic inflammation and glucose intolerance.


Adipose Tissue/enzymology , Encephalitis/enzymology , Glucose Intolerance/enzymology , Hypothalamus/enzymology , I-kappa B Kinase/metabolism , Lung/enzymology , Panniculitis/enzymology , Pneumonia/enzymology , Animals , Aorta/enzymology , Aorta/physiopathology , Blood Glucose/metabolism , Encephalitis/genetics , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Enzyme Activation , Genetic Predisposition to Disease , Glucose Intolerance/blood , Glucose Intolerance/genetics , I-kappa B Kinase/genetics , Insulin Resistance , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/pathology , Mice, Transgenic , Panniculitis/genetics , Phenotype , Pneumonia/genetics , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Brain Behav Immun ; 66: 135-145, 2017 Nov.
Article En | MEDLINE | ID: mdl-28624534

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.


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
16.
Acta Neuropathol Commun ; 5(1): 40, 2017 05 29.
Article En | MEDLINE | ID: mdl-28554330

Onconeural antibodies are associated with cancer and paraneoplastic encephalitis. While their pathogenic role is still largely unknown, their high diagnostic value is undisputed. In this study we describe the discovery of a novel target of autoimmunity in an index case of paraneoplastic encephalitis associated with urogenital cancer.A 75-year-old man with a history of invasive bladder carcinoma 6 years ago with multiple recurrences and a newly discovered renal cell carcinoma presented with seizures and progressive cognitive decline followed by super-refractory status epilepticus. Clinical and ancillary findings including brain biopsy suggested paraneoplastic encephalitis. Immunohistochemistry of the brain biopsy was used to characterize the inflammatory response. Indirect immunofluorescence assay (IFA) was used for autoantibody screening. The autoantigen was identified by histo-immunoprecipitation and mass spectrometry and was validated by expressing the recombinant antigen in HEK293 cells and neutralization tests. Sera from 125 control patients were screened using IFA to test for the novel autoantibodies.IFA analysis of serum revealed a novel autoantibody against brain tissue. An intracellular enzyme, Rho-associated protein kinase 2 (ROCK2), was identified as target-antigen. ROCK2 was expressed in affected brain tissue and archival bladder tumor samples of this patient. Brain histopathology revealed appositions of cytotoxic CD8+ T cells on ROCK2-positive neurons. ROCK2 antibodies were not found in the sera of 20 patients with bladder cancer and 17 with renal cancer, both without neurological symptoms, 49 healthy controls, and 39 patients with other antineuronal autoantibodies. In conclusion, novel onconeural antibodies targeting ROCK2 are associated with paraneoplastic encephalitis and should be screened for when paraneoplastic neurological syndromes, especially in patients with urogenital cancers, occur.


Autoantibodies/immunology , Autoimmune Diseases of the Nervous System/enzymology , Encephalitis/enzymology , Encephalitis/immunology , Paraneoplastic Syndromes, Nervous System/enzymology , Paraneoplastic Syndromes, Nervous System/immunology , rho-Associated Kinases/immunology , Aged , Autoantibodies/blood , Autoimmune Diseases of the Nervous System/immunology , Autoimmunity , Brain/enzymology , Brain/immunology , Carcinoma/immunology , HEK293 Cells , Humans , Kidney Neoplasms/immunology , Male , Urinary Bladder Neoplasms/immunology
17.
Biol Pharm Bull ; 40(5): 557-563, 2017.
Article En | MEDLINE | ID: mdl-28458341

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.


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
18.
CNS Neurosci Ther ; 23(4): 350-359, 2017 Apr.
Article En | MEDLINE | ID: mdl-28191736

AIMS: It has been demonstrated that neuroinflammation is associated with cardiovascular dysfunction. The phosphoinositide-3 kinase (PI3K) signaling in the rostral ventrolateral medulla (RVLM), a key region for sympathetic outflow, is upregulated and contributes to increased blood pressure (BP) and sympathetic outflow in hypertension. This study was designed to determine the role of the PI3K signaling in neuroinflammation in the RVLM of hypertension. METHODS: The normotensive WKY rats were performed by intracisternal infusion of lipopolysaccharide (LPS) or angiotensin II (Ang II) for inducing neuroinflammation. Elisa was used to determine the level of proinflammatory cytokines. Western blot was employed to detect the protein expression of PI3K signaling pathway. Gene silencing of PI3K p110δ subunit and overexpression of angiotensin-converting enzyme 2 (ACE2) were realized by injecting related lentivirus into the RVLM. RESULTS: In the spontaneously hypertensive rats (SHR), the PI3K signaling in the RVLM was upregulated compared with WKY, gene silencing of PI3K in the RVLM significantly reduced BP and renal sympathetic nerve activity (RSNA), but also decreased the levels of proinflammatory cytokines. In the WKY rats, central infusion of LPS and Ang II significantly elevated BP and RSNA, but also increased the levels of proinflammatory cytokines and PI3K signaling activation in the RVLM. These changes in the Ang II-induced hypertension were effectively prevented by gene silencing of PI3K in the RVLM. Furthermore, overexpression of ACE2 in the RVLM significantly attenuated high BP and neuroinflammation, as well as decreased the activation of PI3K signaling in hypertensive rats. CONCLUSION: This study suggests that the PI3K signaling in the RVLM is involved in neuroinflammation in hypertension and plays an important role in the renin-angiotensin system-mediated changes in neuroinflammation in the RVLM.


Encephalitis/enzymology , Hypertension/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Angiotensin II/toxicity , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Encephalitis/etiology , Heart Rate/drug effects , Humans , Hypertension/complications , Lipopolysaccharides/toxicity , Male , Medulla Oblongata/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Phosphatidylinositol 3-Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects , Sympathetic Nervous System/drug effects
19.
Neuropharmacology ; 113(Pt A): 137-147, 2017 02.
Article En | MEDLINE | ID: mdl-27712995

Neuropathic pain, a debilitating pain condition and the underlying pathogenic mechanisms are complex and interwoven amongst each other and still there is scant information available regarding therapies which promise to treat the condition. Evidence indicate that oxidative/nitrosative stress induced poly (ADP-ribose) polymerase (PARP) overactivation initiate neuroinflammation and bioenergetic crisis culminating into neurodegenerative changes following nerve injury. Hence, we investigated the therapeutic effect of combining an antioxidant, quercetin and a PARP inhibitor, 4-amino 1, 8-naphthalimide (4-ANI) on the hallmark deficits induced by chronic constriction injury (CCI) of sciatic nerve in rats. Quercetin (25 mg/kg, p.o.) and 4-ANI (3 mg/kg, p.o.) were administered either alone or in combination for 14 days to examine sciatic functional index, allodynia and hyperalgesia using walking track analysis, Von Frey, acetone spray and hot plate tests respectively. Malondialdehyde, nitrite and glutathione levels were estimated to detect oxidative/nitrosative stress; mitochondrial membrane potential and cytochrome c oxidase activity to assess mitochondrial function; NAD & ATP levels to examine the bioenergetic status and levels of inflammatory markers were evaluated in ipsilateral sciatic nerve. Quercetin and 4-ANI alone improved the pain behaviour and biochemical alterations but the combination therapy demonstrated an appreciable reversal of CCI-induced changes. Nitrotyrosine and Poly ADP-Ribose (PAR) immunopositivity was decreased and nuclear factor erythroid 2-related factor (Nrf-2) levels were increased significantly in micro-sections of the sciatic nerve and dorsal root ganglion (DRG) of treatment group. These results suggest that simultaneous inhibition of oxidative stress-PARP activation cascade may potentially be useful strategies for management of trauma induced neuropathic pain.


1-Naphthylamine/analogs & derivatives , Antioxidants/administration & dosage , Encephalitis/prevention & control , Naphthalimides/administration & dosage , Neuralgia/prevention & control , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Quercetin/administration & dosage , Quinolones/administration & dosage , 1-Naphthylamine/administration & dosage , 1-Naphthylamine/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Antioxidants/therapeutic use , Encephalitis/complications , Encephalitis/enzymology , Hyperalgesia/prevention & control , Male , Mitochondria/drug effects , Mitochondria/metabolism , NAD/metabolism , Naphthalimides/therapeutic use , Neuralgia/complications , Neuralgia/enzymology , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Quercetin/therapeutic use , Quinolones/therapeutic use , Rats , Rats, Sprague-Dawley , Sciatic Nerve/injuries
20.
CNS Neurosci Ther ; 22(8): 661-9, 2016 08.
Article En | MEDLINE | ID: mdl-27156691

BACKGROUND: Microglia-mediated inflammation may play an important role in the pathophysiology progression of neurodegenerative diseases, such as Parkinson's disease (PD), but the molecular mechanisms are poorly understood. AIMS: This study sought to determine whether E3 ubiquitin ligase c-Cbl plays a role in the brain inflammation and to explore the relevant molecular mechanism. METHODS: After BV2 microglial cells and c-Cbl-deficient mice were treated with lipopolysaccharide (LPS), neuroinflammation and microglial activation were evaluated by immunohistochemistry, ELISA and Western blot. We further investigated the possible mechanism of c-Cbl in regulating microglial activation. RESULTS: Here, we showed that the E3 ubiquitin ligase c-Cbl had high expression in brain tissues including substantia nigra pars compacta (SNc), striatum and hippocampus, and it was abundantly expressed in microglia. Systemic LPS administration resulted in more severe microglial activation in CNS and increased expression of brain proinflammatory factors (TNF-α, IL-6, IL-1ß and MCP-1) in c-Cbl knockout mice than wild type mice (WT). Downregulation of c-Cbl expression with c-Cbl siRNA in BV-2 microglial cells demonstrated a more robust increase in the proinflammatory factors release and NF-κB p65 nuclear translocation than that in control siRNA. Interestingly, Akt phosphorylation induced by LPS was also significantly augmented after c-Cbl knockdown. Moreover, blockade of PI3K/Akt activation by LY294002 significantly reduced inflammation response and NF-κB p65 nuclear translocation. CONCLUSION: In sum, c-Cbl inhibits expression of LPS-stimulated proinflammatory cytokines and chemokines in microglia. We demonstrate an unprecedented role for c-Cbl in microglia-mediated neuroinflammation involving PI3K/Akt/NF-κB pathway.


Encephalitis/enzymology , Microglia/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-cbl/deficiency , Signal Transduction/physiology , Animals , Brain/metabolism , Brain/pathology , Cell Line, Transformed , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/etiology , Encephalitis/pathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Microglia/drug effects , NF-kappa B/metabolism , Nitrites/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
...