Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Parasite Immunol ; 45(12): e13014, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37807942

ABSTRACT

This study investigated a 'de Novo' medicinal herb, Ferula asafetida (FA), against toxoplasma encephalitis either alone or combined with spiramycin (SP). Female Swiss-Webster mice (n = 72) were divided into three batches. Batch-I received no DMS to serve as an immunocompetent control, batch-II was immune-suppressed with the DMS (0.25 mg/g/day) for 14 days pre-infection, whilst batch-III was immune-suppressed with the DMS on the same day of infection. All experimental mice were inoculated with Toxoplasma gondii ME49 cysts (n = 75). Each batch was split into four subgroups: Mono-SP, mono-FA, combined drug (SP + FA), or neither. Therapies were administered on day zero of infection in batches (I and II) and 35 days post-infection in batch (III). Treatments lasted for 14 days, and mice were sacrificed 60 days post-infection. Histopathological changes, cysts load, and CD4 and CD8 T-cells were counted in brain tissues. The cyst-load count in mice receiving SP + FA was significantly (p < .0001) the least compared to the mono treatments in all protocols. Interestingly, the combined therapy demolished the T-cell subsets to zero in immunocompetent and immunocompromised infected mice. In conclusion, F. asafetida might be a powerfully natural, safe vehicle of SP in the digestive system and/or across the brain-blood barrier to control toxoplasmosis even through immunodeficient conditions.


Subject(s)
Encephalitis , Ferula , Spiramycin , Toxoplasma , Toxoplasmosis, Animal , Toxoplasmosis, Cerebral , Female , Mice , Animals , Spiramycin/therapeutic use , Brain , Toxoplasmosis, Animal/drug therapy , Encephalitis/drug therapy , Encephalitis/pathology
2.
Arch Biochem Biophys ; 740: 109598, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37054769

ABSTRACT

Inflammation and oxidative stress are mechanisms which potentially underlie the brain damage that can occur after cardiac ischemic and reperfusion (I/R) injury. 2i-10 is a new anti-inflammatory agent, acting via direct inhibition of myeloid differentiation factor 2 (MD2). However, the effects of 2i-10 and the antioxidant N-acetylcysteine (NAC) on pathologic brain in cardiac I/R injury are unknown. We hypothesized that 2i-10 and NAC offer similar neuroprotection levels against dendritic spine reduction through attenuation of brain inflammation, loss of tight junction integrity, mitochondrial dysfunction, reactive gliosis, and suppression of AD protein expression in rats with cardiac I/R injury. Male rats were allocated to either sham or acute cardiac I/R group (30 min of cardiac ischemia and 120 min of reperfusion). Rats in cardiac I/R group were given one of following treatments intravenously at the onset of reperfusion: vehicle, 2i-10 (20 or 40 mg/kg), and NAC (75 or 150 mg/kg). The brain was then used to determine biochemical parameters. Cardiac I/R led to cardiac dysfunction with dendritic spine loss, loss of tight junction integrity, brain inflammation, and mitochondrial dysfunction. Treatment with 2i-10 (both doses) effectively reduced cardiac dysfunction, tau hyperphosphorylation, brain inflammation, mitochondrial dysfunction, dendritic spine loss, and improved tight junction integrity. Although both doses of NAC effectively reduced brain mitochondrial dysfunction, treatment using a high dose of NAC reduced cardiac dysfunction, brain inflammation, and dendritic spine loss. In conclusion, treatment with 2i-10 and a high dose of NAC at the onset of reperfusion alleviated brain inflammation and mitochondrial dysfunction, consequently reducing dendritic spine loss in rats with cardiac I/R injury.


Subject(s)
Encephalitis , Myocardial Reperfusion Injury , Reperfusion Injury , Rats , Male , Animals , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Brain/metabolism , Oxidative Stress , Encephalitis/pathology , Ischemia/pathology
4.
Nutrients ; 13(10)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34684462

ABSTRACT

Recent studies have shown a rise in precocious puberty, especially in girls. At the same time, childhood obesity due to overnutrition and energy imbalance is rising too. Nutrition and fertility are currently facing major challenges in our societies, and are interconnected. Studies have shown that high-fat and/or high-glycaemic-index diet can cause hypothalamic inflammation and microglial activation. Molecular and animal studies reveal that microglial activation seems to produce and activate prostaglandins, neurotrophic factors activating GnRH (gonadotropin-releasing hormone expressing neurons), thus initiating precocious puberty. GnRH neurons' mechanisms of excitability are not well understood. In this review, we study the phenomenon of the rise of precocious puberty, we examine the physiology of GnRH neurons, and we review the recent literature regarding the pathophysiological mechanisms that connect diet-induced hypothalamic inflammation and diet-induced phoenixin regulation with precocious puberty.


Subject(s)
Diet/adverse effects , Encephalitis/complications , Hypothalamus/metabolism , Peptide Hormones/metabolism , Puberty, Precocious/etiology , Puberty, Precocious/metabolism , Animals , Biomarkers , Disease Susceptibility , Encephalitis/etiology , Encephalitis/pathology , Humans , Hypothalamus/pathology
5.
Life Sci ; 286: 119989, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34597609

ABSTRACT

AIMS: Isoformononetin (IFN), a methoxyl isoflavone present in most of human dietary supplements. However, being a highly potent antioxidant and anti-inflammatory molecule, its activity against neuronal oxidative stress and neuroinflammation has not been explored till now. The present study was inquested to assess the antioxidant, anti-apoptotic and anti-inflammatory activity of IFN against streptozotocin induced neuroinflammation in different brain regions of rat. MAIN METHODS: Four groups of animals were subjected to treatment as control, toxic control (STZ; single intracerebrovascular injection), third group (STZ + IFN; 20 mg/kg p.o.), fourth group (IFN) for 14 days. The different brain regions of rats were evaluated for inflammatory, apoptotic and biochemical antioxidant markers. The brain tissues were further assessed for gene expression, immunohistochemical and western blotting examination for localization of inflammasome cascade expression that plays a pivotal role in neuroinflammation. KEY FINDINGS: The modulation in oxidant/antioxidant status after exposure of STZ was significantly balanced after administration of IFN to rats. Further, IFN was also found to be an apoptotic agent as it modulates the apoptotic gene (Bax) and anti-apoptotic gene (BcL2) expression. IFN significantly curtailed the augmented protein expression of NLRP3, NLRP2, ASC, NFκBP65, IL-1ß and caspase-1 due to STZ administration in cortex and hippocampus rat brain regions. SIGNIFICANCE: The aforementioned results proclaim the neuroprotective functioning of IFN against STZ induced inflammation. IFN significantly prevents the neuroinflammation by decreasing the generation of ROS that reduces the activation of NLRP3/ASC/IL-1 axis thereby exerting neuroprotection as evidenced in rat model of STZ induced neuroninflammation.


Subject(s)
Antioxidants/pharmacology , CARD Signaling Adaptor Proteins/metabolism , Encephalitis/prevention & control , Interleukin-1/metabolism , Isoflavones/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Streptozocin/toxicity , Animals , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/pathology , Gene Expression/physiology , Interferons/physiology , Lipid Peroxidation/drug effects , Nitric Oxide/biosynthesis , Oxidative Stress/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
6.
Am J Respir Crit Care Med ; 204(12): 1391-1402, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34491883

ABSTRACT

Rationale: Mechanical ventilation (MV) is associated with hippocampal apoptosis and inflammation, and it is important to study strategies to mitigate them. Objectives: To explore whether temporary transvenous diaphragm neurostimulation (TTDN) in association with MV mitigates hippocampal apoptosis and inflammation after 50 hours of MV. Methods: Normal-lung porcine study comparing apoptotic index, inflammatory markers, and neurological-damage serum markers between never-ventilated subjects, subjects undergoing 50 hours of MV plus either TTDN every other breath or every breath, and subjects undergoing 50 hours of MV (MV group). MV settings in volume control were Vt of 8 ml/kg, and positive end-expiratory pressure of 5 cm H2O. Measurements and Main Results: Apoptotic indices, microglia percentages, and reactive astrocyte percentages were greater in the MV group in comparison with the other groups (P < 0.05). Transpulmonary pressure at baseline and at study end were both lower in the group receiving TTDN every breath, but lung injury scores and systemic inflammatory markers were not different between the groups. Serum concentrations of four neurological-damage markers were lower in the group receiving TTDN every breath than in the MV group (P < 0.05). Heart rate variability declined significantly in the MV group and increased significantly in both TTDN groups over the course of the experiments. Conclusions: Our study found that mechanical ventilation is associated with hippocampal apoptosis and inflammation, independent of lung injury and systemic inflammation. Also, in a porcine model, TTDN results in neuroprotection after 50 hours, and the degree of neuroprotection increases with greater exposure to TTDN.


Subject(s)
Apoptosis , Brain Injuries/prevention & control , Diaphragm/innervation , Electric Stimulation Therapy/methods , Encephalitis/prevention & control , Hippocampus/pathology , Respiration, Artificial/adverse effects , Animals , Brain Injuries/diagnosis , Brain Injuries/etiology , Brain Injuries/pathology , Encephalitis/diagnosis , Encephalitis/etiology , Encephalitis/pathology , Female , Phrenic Nerve , Respiration, Artificial/methods , Swine , Treatment Outcome
7.
Stroke ; 52(2): 687-698, 2021 01.
Article in English | MEDLINE | ID: mdl-33412903

ABSTRACT

BACKGROUND AND PURPOSE: Stroke disrupts neuronal functions in both local and remotely connected regions, leading to network-wide deficits that can hinder recovery. The thalamus is particularly affected, with progressive development of neurodegeneration accompanied by inflammatory responses. However, the complexity of the involved inflammatory responses is poorly understood. Herein we investigated the spatiotemporal changes in the secondary degenerative thalamus after cortical stroke, using targeted transcriptome approach in conjunction with histology and flow cytometry. METHODS: Cortical ischemic stroke was generated by permanent occlusion of the left middle cerebral artery in male C57BL6J mice. Neurodegeneration, neuroinflammatory responses, and microglial activation were examined in naive and stroke mice at from poststroke days (PD) 1 to 84, in both ipsilesional somatosensory cortex and ipsilesional thalamus. NanoString neuropathology panel (780 genes) was used to examine transcriptome changes at PD7 and PD28. Fluorescence activated cell sorting was used to collect CD11c+ microglia from ipsilesional thalamus, and gene expressions were validated by quantitative real-time polymerase chain reaction. RESULTS: Neurodegeneration in the thalamus was detected at PD7 and progressively worsened by PD28. This was accompanied by rapid microglial activation detected as early as PD1, which preceded the neurodegenerative changes. Transcriptome analysis showed higher number of differentially expressed genes in ipsilesional thalamus at PD28. Notably, neuroinflammation was the top activated pathway, and microglia was the most enriched cell type. Itgax (CD11c) was the most significantly increased gene, and its expression was highly detected in microglia. Flow-sorted CD11c+ microglia from degenerative thalamus indicated molecular signatures similar to neurodegenerative disease-associated microglia; these included downregulated Tmem119 and CX3CR1 and upregulated ApoE, Axl, LpL, CSF1, and Cst7. CONCLUSIONS: Our findings demonstrate the dynamic changes of microglia after stroke and highlight the importance of investigating stroke network-wide deficits. Importantly, we report the existence of a unique subtype of microglia (CD11c+) with neurodegenerative disease-associated microglia features in the degenerative thalamus after stroke.


Subject(s)
Cerebral Cortex/pathology , Microglia/pathology , Neurodegenerative Diseases/pathology , Stroke/complications , Stroke/pathology , Thalamic Diseases/etiology , Thalamic Diseases/pathology , Animals , CD11 Antigens/chemistry , Cerebrovascular Circulation , Encephalitis/pathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Somatosensory Cortex/pathology , Thalamus/pathology , Transcriptome
8.
Neurochem Int ; 144: 104931, 2021 03.
Article in English | MEDLINE | ID: mdl-33276023

ABSTRACT

INTRODUCTION: The accumulation of oxidative stress, neuroinflammation and abnormal aggregation of amyloid ß-peptide (Aß) have been shown to induce synaptic dysfunction and memory deficits in Alzheimer's disease (AD). Cellular depletion of the major endogenous antioxidant Glutathione (GSH) has been linked to cognitive decline and the development of AD pathology. Supplementation with γ-glutamylcysteine (γ-GC), the immediate precursor and the limiting substrate for GSH biosynthesis, can transiently augment cellular GSH levels by bypassing the regulation of GSH homeostasis. METHODS: In the present study, we investigated the effect of dietary supplementation of γ-GC on oxidative stress and Aß pathology in the brains of APP/PS1 mice. The APP/PS1 mice were fed γ-GC from 3 months of age with biomarkers of apoptosis and cell death, oxidative stress, neuroinflammation and Aß load being assessed at 6 months of age. RESULTS: Our data showed that supplementation with γ-GC lowered the levels of brain lipid peroxidation, protein carbonyls and apoptosis, increased both total GSH and the glutathione/glutathione disulphide (GSH/GSSG) ratio and replenished ATP and the activities of the antioxidant enzymes (superoxide dismutase (SOD), catalase, glutamine synthetase and glutathione peroxidase (GPX)), the latter being a key regulator of ferroptosis. Brain Aß load was lower and acetylcholinesterase (AChE) activity was markedly improved compared to APP/PS1 mice fed a standard chow diet. Alteration in brain cytokine levels and matrix metalloproteinase enzymes MMP-2 and MMP-9 suggested that γ-GC may lower inflammation and enhance Aß plaque clearance in vivo. Spatial memory was also improved by γ-GC as determined using the Morris water maze. CONCLUSION: Our data collectively suggested that supplementation with γ-GC may represent a novel strategy for the treatment and/or prevention of cognitive impairment and neurodegeneration.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid/antagonists & inhibitors , Dipeptides/administration & dosage , Encephalitis/drug therapy , Oxidative Stress/drug effects , Spatial Memory/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Animals , Dietary Supplements , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Male , Mice , Mice, Transgenic , Oxidative Stress/physiology , Spatial Memory/physiology
9.
BMC Neurol ; 20(1): 125, 2020 Apr 08.
Article in English | MEDLINE | ID: mdl-32268889

ABSTRACT

BACKGROUND: Neuronal intranuclear inclusion disease (NIID) is a rare neurodegenerative condition characterized by the loss of neurons and the presence of eosinophilic nuclear inclusions in the central and peripheral nervous system, skin and visceral organs. In this paper, we present a case of NIID with recurrent encephalitic attacks that remained stable and nonprogressive for seven years; no such case has previously been reported. CASE PRESENTATION: A 63-year-old female was hospitalized due to light-headedness, vomiting, unstable gait and cognitive impairment. Seven years prior, she had experienced an episode of light-headedness, central facial paralysis, unstable gait, aphasia, nausea, vomiting and loss of consciousness. She regained consciousness within 12 h, and her other symptoms were completely resolved within one week. During the present hospitalization, a brain magnetic resonance imaging (MRI) examination detected high signal intensity on diffusion-weighted imaging (DWI) of the bilateral frontal grey matter-white matter junction. We reviewed the patient's previous MRI results and found that she had also had high signal intensity on DWI of the bilateral frontal grey matter-white matter junction seven years prior. In the intervening seven years, the high signal intensity in the frontal lobes had spread along the grey matter-white matter junction, but the deep white matter remained unaffected. Skin biopsy was performed, and intranuclear inclusions were found in adipocytes, fibroblasts and sweat gland cells. GGC repeat expansions in the NOTCH2NLC (Notch 2 N-terminal like C) gene confirmed the diagnosis of NIID. She received supportive treatment such as nutrition support therapy and vitamin B and C supplementation, as well as symptomatic treatment during hospitalization. The patient's symptoms were completely relieved within one week. CONCLUSION: This is a detailed report of a case of NIID with multiple reversible encephalitic attacks, diagnosed by clinical symptoms, intranuclear inclusions, characteristic DWI signals, and genetic tests.


Subject(s)
Encephalitis/pathology , Neurodegenerative Diseases/diagnosis , Biopsy , Cognitive Dysfunction/pathology , Diffusion Magnetic Resonance Imaging , Female , Humans , Intranuclear Inclusion Bodies , Magnetic Resonance Imaging , Middle Aged , Skin/pathology
10.
J Neuroendocrinol ; 31(8): e12757, 2019 08.
Article in English | MEDLINE | ID: mdl-31233242

ABSTRACT

The early-life environment is important in programming brain development, and metabolic disruptions at this time can have long-lasting effects. Previously, we have shown that rats overfed for the first 3 weeks of their neonatal life maintain obesity into adulthood. Neonatal overfeeding also leads to primed hypothalamic and hippocampal microglia that are hyper-responsive to an immune challenge in adulthood. However, whether this microglial priming contributes to the obese phenotype and whether it is possible to reverse either the obesity or the microglial priming are not clear. In the present study, we hypothesised that an intervention with minocycline during the juvenile period (postnatal day 21-42) would normalise both the microglial priming and obesity. To induce obesity in neonatal Wistar rats, we manipulated the litter sizes in which they were suckled, yielding litters of 12 (control-fed) or four (neonatally overfed). After weaning, we administered minocycline i.p. every second day for a 3-week period and examined body composition and microglial profiles 24 hours following an immune challenge with lipopolysaccharide. As demonstrated previously, neonatal overfeeding resulted in prolonged weight gain. However, minocycline failed to reverse this effect. Minocycline did reverse microglial priming in feeding-related regions of the hypothalamus, with minimal effects on pro-inflammatory cytokines and on microglial number and morphology in the hippocampus. Thus, the programming effect of neonatal overfeeding on microglial priming can be ameliorated by minocycline later in life. However, the persistent obesity seen after neonatal overfeeding is likely not driven by changes in hypothalamic inflammation and microglial activity.


Subject(s)
Encephalitis/physiopathology , Hypothalamus/pathology , Microglia/physiology , Obesity/etiology , Overnutrition/complications , Animals , Animals, Newborn , Cellular Reprogramming/drug effects , Encephalitis/complications , Encephalitis/pathology , Female , Hypothalamus/drug effects , Male , Microglia/drug effects , Microglia/pathology , Minocycline/pharmacology , Obesity/pathology , Obesity/physiopathology , Overnutrition/pathology , Overnutrition/physiopathology , Pregnancy , Rats , Rats, Wistar , Weight Gain/drug effects , Weight Gain/physiology
11.
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
12.
Phytother Res ; 32(12): 2531-2540, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30277284

ABSTRACT

The acute exposure of trimethyltin (TMT) develops clinical syndrome characterized by amnesia, aggressive behavior, and complex seizures. This neurotoxicant selectively induces hippocampal neuronal injury and glial activation accompanied with resultant neuroinflammation. Here we report two candidates ginsenosides Rg3 and Rh2 as neuroprotection agents using a mouse model of TMT intoxication via a single injection (2 mg/kg) and primary neuronal culture systems. Four-week administration of Rg3 or Rh2 significantly reduced TMT-induced seizures and behavioral changes. Rg3 and Rh2 significantly attenuated the oxidative stress evidenced by improvement on antioxidant enzymes and neuronal loss and astrocytic activation in mouse brain. In primary cultures, TMT induced significant neuronal death after 24-h intoxication and vigorous secretion of inflammatory cytokines (IL-1α/ß, IL-6, TNF-α, and MCP-1) in astrocytes. Pretreatment with Rg3 or Rh2 not only reduced cell death but efficiently suppressed above mentioned inflammatory cytokines confirmed by antibody array test. The underlying protective mechanism by Rg3 and Rh2 was delineated through selective upregulation of PI3K/Akt and suppression of ERK activation. Intriguingly, Rg3 and Rh2 protected oligodendrocyte progenitor cells (O-2A) from TMT intoxication via promoting type 2 astrocytic differentiation without further inflammatory activation. Collectively, Rg3 and Rh2 interventions aimed at reducing oxidative stress and neuroinflammation neurotoxicity therefore are of therapeutic benefit in TMT-induced neurodegeneration.


Subject(s)
Encephalitis/prevention & control , Ginsenosides/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Embryo, Mammalian , Encephalitis/chemically induced , Encephalitis/pathology , Neurons/physiology , Neuroprotection/drug effects , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Trimethyltin Compounds
13.
J Neuroinflammation ; 15(1): 299, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30373627

ABSTRACT

BACKGROUND: Although studies have reported an increased risk for mood disorders in Hashimoto's thyroiditis (HT) patients even in the euthyroid state, the mechanisms involved remain unclear. Neuroinflammation may play a key role in the etiology of mood disorders in humans and behavioral disturbances in rodents. Therefore, this study established a euthyroid HT model in mice and investigated whether HT itself was capable of triggering neuroinflammation accompanied by emotional alterations. METHODS: Experimental HT was induced by immunizing NOD mice with thyroglobulin and adjuvant twice. Four weeks after the last challenge, mice were tested for anxiety-like behavior in the open field and elevated plus maze tests and depression-like behavior in the forced swimming and tail suspension tests. Then, animals were sacrificed for thyroid-related parameter measure as well as detection of cellular and molecular events associated with neuroinflammation. The changes in components of central serotonin signaling were also investigated. RESULTS: HT mice showed intrathyroidal monocyte infiltration and rising serum thyroid autoantibody levels accompanied by normal thyroid function, which defines euthyroid HT in humans. These mice displayed more anxiety- and depressive-like behaviors than controls. HT mice further showed microglia and astrocyte activation in the frontal cortex detected by immunohistochemistry, real-time RT-PCR, and transmission electron microscopy (TEM). These observations were also accompanied by enhanced gene expression of proinflammatory cytokines IL-1ß and TNF-α in the frontal cortex. Despite this inflammatory response, no signs of neuronal apoptosis were visible by the TUNEL staining and TEM in the frontal cortex of HT mice. Additionally, IDO1 and SERT, key serotonin-system-related genes activated by proinflammatory cytokines, were upregulated in HT mice, accompanied by reduced frontal cortex serotonin levels. CONCLUSIONS: Our results are the first to suggest that HT induces neuroinflammation and alters related serotonin signaling in the euthyroid state, which may underlie the deleterious effects of HT itself on emotional function.


Subject(s)
Affective Symptoms/etiology , Encephalitis/etiology , Hashimoto Disease/complications , Animals , Brain/pathology , Brain/ultrastructure , Calcium-Binding Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/pathology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Freund's Adjuvant/toxicity , Glial Fibrillary Acidic Protein/metabolism , Hashimoto Disease/etiology , Hashimoto Disease/pathology , Hindlimb Suspension , In Situ Nick-End Labeling , Maze Learning/drug effects , Mice , Mice, Inbred NOD , Microfilament Proteins/metabolism , Microscopy, Electron, Transmission , Neuroglia/pathology , Neuroglia/ultrastructure , Neurons/pathology , Neurons/ultrastructure , Swimming/psychology
14.
Biomed Pharmacother ; 106: 1153-1160, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119182

ABSTRACT

Gypenosides, a saponins extract isolated from the Gynostemma pentaphyllum plant, produces neuroprotective effects in the brain. Our previous studies have shown that hippocampal glucocorticoid receptor (GR)-brain-derived neurotrophic factor (BDNF)-TrkB signaling was involved in the antidepressant-like effects of gypenosides. It remains unknown whether gypenosides could alleviate neuroinflammation in depressive-like animals. The aim of the present study was to address this issue in chronic unpredictable mild stress (CUMS). Gypenosides was administrated for four weeks, followed by sucrose preference test and tail suspension test, which were performed to evaluate the effects of gypenosides. The results showed that gypenosides reversed both the decreased sucrose preference and increased immobility time in CUMS mice. In addition, gypenosides also attenuated the increase of pro-inflammatory cytokine levels in the hippocampus of CUMS animals. Furthermore, the activation of NF-κB, as well as its upstream mediators IKKα and IKKß were inhibited by gypenosides. Last but not the least, CUMS promoted the activation of microglia, while gypenosides suppressed it according to the reduced number of iba1 positive cells. In conclusion, this study demonstrates that gypenosides exhibits the antidepressant-like effects in mice, which may be mediated by the inhibition of microglia and NF-κB signaling in the hippocampus.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Depression/drug therapy , Encephalitis/prevention & control , Hippocampus/drug effects , Stress, Psychological/drug therapy , Animals , Calcium-Binding Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Depression/metabolism , Depression/pathology , Depression/psychology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Encephalitis/psychology , Feeding Behavior/drug effects , Gynostemma , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Activity/drug effects , NF-kappa B/metabolism , Phosphorylation , Plant Extracts/pharmacology , Signal Transduction/drug effects , Stress, Psychological/metabolism , Stress, Psychological/pathology , Stress, Psychological/psychology
15.
CNS Neurol Disord Drug Targets ; 17(9): 680-688, 2018.
Article in English | MEDLINE | ID: mdl-29938622

ABSTRACT

BACKGROUND AND OBJECTIVE: A steep rise in the incidences of neurodegenerative disorders could be the combined effect of several non-genetic factors such as increased life expectancy, environmental pollutants, lifestyle, and dietary habits, as population-level genetic change require multiple generations. Emerging evidence suggests that chronic over-nutrition induces brain metabolic stress and neuroinflammation, and are individually known to promote neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Although the association of metabolic disorders such as diabetes, hypertension, dyslipidemia, and atherosclerosis with the dietary habits is well known, neuronal implications of diet and nutritional factors is still in its infancy. Transcriptomics and proteomics-based studies support the view that nutraceuticals target multiple neuroprotective pathways in a slow but effective manner without causing severe adverse effects, and may represent the future of tackling neurodegenerative disorders. CONCLUSION: In this article we i) review the diet/dietary supplement connection with brain metabolic stress and neuroinflammation and ii) summarize current knowledge of the effects of nutraceuticals on neurodegenerative disorders.


Subject(s)
Brain/physiopathology , Dietary Supplements , Encephalitis/pathology , Nutrients/metabolism , Stress, Physiological/physiology , Animals , Brain/metabolism , Encephalitis/therapy , Humans
16.
Nutrients ; 9(10)2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28961195

ABSTRACT

Choline is involved in relevant neurochemical processes. In particular, it is the precursor and metabolite of acetylcholine (ACh). Choline is an essential component of different membrane phospholipids that are involved in intraneuronal signal transduction. On the other hand, cholinergic precursors are involved in ACh release and carry out a neuroprotective effect based on an anti-inflammatory action. Based on these findings, the present study was designed to evaluate the effects of choline and choline precursor (Choline alphoscerate, GPC) in the modulation of inflammatory processes in the rat brain. Male Wistar rats were intraperitoneally treated with 87 mg of choline chloride/kg/day (65 mg/kg/day of choline), and at choline-equivalent doses of GPC (150 mg/kg/day) and vehicle for two weeks. The brains were dissected and used for immunochemical and immunohistochemical analysis. Inflammatory cytokines (Interleukin-1ß, IL-1ß; Interleukin-6 , IL-6 and Tumor Necrosis Factor-α, TNF-α) and endothelial adhesion molecules (Intercellular Adhesion Molecule, ICAM-1 and Vascular cell Adhesion Molecule, VCAM-1) were studied in the frontal cortex, hippocampus, and cerebellum. The results clearly demonstrated that treatment with choline or GPC did not affect the expression of the inflammatory markers in the different cerebral areas evaluated. Therefore, choline and GPC did not stimulate the inflammatory processes that we assessed in this study.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cerebral Cortex/drug effects , Choline/therapeutic use , Encephalitis/prevention & control , Glycerylphosphorylcholine/therapeutic use , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Biomarkers/metabolism , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Choline/administration & dosage , Choline/adverse effects , Cytokines/metabolism , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/pathology , Frontal Lobe/drug effects , Frontal Lobe/immunology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Glycerylphosphorylcholine/administration & dosage , Glycerylphosphorylcholine/adverse effects , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/pathology , Injections, Intraperitoneal , Intercellular Adhesion Molecule-1/metabolism , Male , Nerve Tissue Proteins/metabolism , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/cerebrospinal fluid , Rats, Wistar , Vascular Cell Adhesion Molecule-1/metabolism
17.
Sci Rep ; 7(1): 3697, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623340

ABSTRACT

Glucokinase (GK), the hexokinase involved in glucosensing in pancreatic ß-cells, is also expressed in arcuate nucleus (AN) neurons and hypothalamic tanycytes, the cells that surround the basal third ventricle (3V). Several lines of evidence suggest that tanycytes may be involved in the regulation of energy homeostasis. Tanycytes have extended cell processes that contact the feeding-regulating neurons in the AN, particularly, agouti-related protein (AgRP), neuropeptide Y (NPY), cocaine- and amphetamine-regulated transcript (CART) and proopiomelanocortin (POMC) neurons. In this study, we developed an adenovirus expressing GK shRNA to inhibit GK expression in vivo. When injected into the 3V of rats, this adenovirus preferentially transduced tanycytes. qRT-PCR and Western blot assays confirmed GK mRNA and protein levels were lower in GK knockdown animals compared to the controls. In response to an intracerebroventricular glucose injection, the mRNA levels of anorexigenic POMC and CART and orexigenic AgRP and NPY neuropeptides were altered in GK knockdown animals. Similarly, food intake, meal duration, frequency of eating events and the cumulative eating time were increased, whereas the intervals between meals were decreased in GK knockdown rats, suggesting a decrease in satiety. Thus, GK expression in the ventricular cells appears to play an important role in feeding behavior.


Subject(s)
Adenoviridae/physiology , Feeding Behavior , Glucokinase/metabolism , Hypothalamus/metabolism , Hypothalamus/physiopathology , Adenoviridae Infections , Animals , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/pathology , Gene Expression , Gene Expression Regulation , Genes, Reporter , Hypothalamus/pathology , Hypothalamus/virology , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
Mol Cell Neurosci ; 80: 123-133, 2017 04.
Article in English | MEDLINE | ID: mdl-28286294

ABSTRACT

An important hallmark of various neurodegenerative disorders is the proliferation and activation of microglial cells, the resident immune cells of the central nervous system (CNS). Mice that lack multifunctional protein-2 (MFP2), the key enzyme in peroxisomal ß-oxidation, develop excessive microgliosis that positively correlates with behavioral deficits whereas no neuronal loss occurs. However, the precise contribution of neuroinflammation to the fatal neuropathology of MFP2 deficiency remains largely unknown. Here, we first attempted to suppress the inflammatory response by administering various anti-inflammatory drugs but they failed to reduce microgliosis. Subsequently, Mfp2-/- mice were treated with the selective colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 as microglial proliferation and survival is dependent on CSF1R signaling. This resulted in the elimination of >95% of microglia from control mice but only 70% of the expanded microglial population from Mfp2-/- mice. Despite microglial diminution in Mfp2-/- brain, inflammatory markers remained unaltered and residual microglia persisted in a reactive state. CSF1R inhibition did not prevent neuronal dysfunction, cognitive decline and clinical deterioration of Mfp2-/- mice. Collectively, the unaltered inflammatory profile despite suppressed microgliosis concurrent with persevering clinical decline strengthens our hypothesis that neuroinflammation importantly contributes to the Mfp2-/- phenotype.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalitis , Gliosis/etiology , Peroxisomal Multifunctional Protein-2/deficiency , Acoustic Stimulation , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Differentiation/metabolism , Avoidance Learning/drug effects , Avoidance Learning/physiology , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Encephalitis/complications , Encephalitis/genetics , Encephalitis/pathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Evoked Potentials, Auditory, Brain Stem/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Muscle Strength/drug effects , Muscle Strength/genetics , Peroxisomal Multifunctional Protein-2/genetics , Severity of Illness Index
19.
J Alzheimers Dis ; 60(s1): S221-S235, 2017.
Article in English | MEDLINE | ID: mdl-28222524

ABSTRACT

BACKGROUND: Experimental studies have demonstrated that aluminum is an environmental toxin that induces neuroinflammation and the development of Alzheimer's disease. OBJECTIVE: In this report, we investigated the beneficial effect of a combination of resveratrol and curcumin to reduce aluminum-induced neuroinflammation. METHOD: We employed both an in vivo model of aluminum-induced neuroinflammation and an in vitro aluminum stimulated cultured PC-12 cells. Neuroinflammation in rats was assessed by measuring the expression of ß-secretase, amyloid-ß protein precursor, and γ-subunits (PS-1 and PS-2), along with the inflammatory COX-2, Il-1ß, Il-1α, and TNF-α. Furthermore, we measured the expression profiles of neuro-protective Apurinic/apyrimidinic endonuclease 1 (APE1) protein and let-7c microRNA. In parallel, PC-12 cells were treated with 0.5 mM aluminum to induce a neuroinflammation-like state. In addition, curcumin effect, as a selective COX-2 expression inhibitor, was detected in a time course manner. RESULTS: An overall significant attenuation of the inflammatory markers, as well as a decrease in the amyloidogenic mediators, was observed in resveratrol-curcumin treated rats. The therapeutic effect was also confirmed by transmission electron microscopic analysis of the brain cortexes. APE1 was significantly induced by resveratrol-curcumin combination. Both in vivo and in vitro studies indicated that Let-7c expression is significantly reduced after aluminum stimulation, an effect that was partially suppressed by co-addition of either resveratrol or curcumin and totally restored to the normal level by their combination. CONCLUSIONS: The present study clearly indicates the synergistic and therapeutic effect of a  resveratrol-curcumin combination. We also show that both compounds exert beneficial effect either cooperatively or through differential molecular mechanisms in counteracting aluminum-induced neuroinflammation.


Subject(s)
Aluminum Compounds/toxicity , Chlorides/toxicity , Curcumin/therapeutic use , Encephalitis/chemically induced , Encephalitis/drug therapy , Neuroprotective Agents/therapeutic use , Stilbenes/toxicity , Acetylcholinesterase/metabolism , Aluminum Chloride , Animals , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Catalase/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Drug Combinations , Encephalitis/pathology , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Male , Oxidative Stress/drug effects , PC12 Cells , Rats , Rats, Wistar , Resveratrol , Superoxide Dismutase/metabolism
20.
Sci Rep ; 7: 41032, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102362

ABSTRACT

The neurologic manifestations of neonatal hyperbilirubinemia in the central nervous system (CNS) exhibit high variations in the severity and appearance of motor, auditory and cognitive symptoms, which is suggestive of a still unexplained selective topography of bilirubin-induced damage. By applying the organotypic brain culture (OBC: preserving in vitro the cellular complexity, connection and architecture of the in vivo brain) technique to study hyperbilirubinemia, we mapped the regional target of bilirubin-induced damage, demonstrated a multifactorial toxic action of bilirubin, and used this information to evaluate the efficacy of drugs applicable to newborns to protect the brain. OBCs from 8-day-old rat pups showed a 2-13 fold higher sensitivity to bilirubin damage than 2-day-old preparations. The hippocampus, inferior colliculus and cerebral cortex were the only brain regions affected, presenting a mixed inflammatory-oxidative mechanism. Glutamate excitotoxicity was appreciable in only the hippocampus and inferior colliculus. Single drug treatment (indomethacin, curcumin, MgCl2) significantly improved cell viability in all regions, while the combined (cocktail) administration of the three drugs almost completely prevented damage in the most affected area (hippocampus). Our data may supports an innovative (complementary to phototherapy) approach for directly protecting the newborn brain from bilirubin neurotoxicity.


Subject(s)
Bilirubin/toxicity , Brain Diseases, Metabolic/pathology , Brain/drug effects , Brain/pathology , Encephalitis/pathology , Hyperbilirubinemia/complications , Animals , Cell Survival/drug effects , Curcumin/pharmacology , Indomethacin/pharmacology , Inflammation/pathology , Magnesium Chloride/pharmacology , Models, Biological , Neuroprotective Agents/pharmacology , Organ Culture Techniques , Oxidative Stress , Rats
SELECTION OF CITATIONS
SEARCH DETAIL