Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 484
1.
J Chem Neuroanat ; 137: 102402, 2024 Apr.
Article En | MEDLINE | ID: mdl-38428651

Curcumin, a bioactive polyphenol derived from turmeric, has been reported to have anti-inflammatory properties. The current study investigated the anti-inflammatory effect of curcumin in the hippocampal subfields (CA1 and CA3) after exposure to cobalt (Co) and the impact of ERK protein. Twenty-eight albino Wistar rats were divided into four groups, each with seven randomly selected rats as follows: Control (distilled water), Cobalt (Co) only (40 mg/kg), 120 mg/kg or 240 mg/kg curcumin + Co (40 mg/kg). Treatment was via oral gavage for 28 days. We performed a biochemical investigation to determine the levels of proinflammatory cytokines (TNFα and IL-1ß). Furthermore, we conducted an immunohistochemical evaluation to assess the expression of IBA1 by microglial cells and the immunoexpression of ERK protein in the hippocampus. Results revealed a significant (p<0.05) elevation in the tissue level of TNFα and IL-1ß, an increase in the number of IBA1-positive microglia, and upregulation of ERK protein in the hippocampal subfields of the rats after exposure to cobalt-only. Nevertheless, pretreatment with curcumin restored these parameters to levels comparable to control. In conclusion, our results showed that curcumin abrogated the Co-induced neuroinflammation by suppressing the release of proinflammatory biomarkers, reducing microgliosis, and modulating the ERK/MAPK pathway.


Cobalt , Curcumin , Cytokines , Hippocampus , MAP Kinase Signaling System , Microglia , Neuroinflammatory Diseases , Rats, Wistar , Animals , Curcumin/pharmacology , Cobalt/toxicity , Rats , MAP Kinase Signaling System/drug effects , Male , Cytokines/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/chemically induced , Microglia/drug effects , Microglia/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Gliosis/metabolism , Gliosis/chemically induced , Gliosis/drug therapy
2.
Sci Rep ; 13(1): 22712, 2023 12 19.
Article En | MEDLINE | ID: mdl-38123648

Retinal Müller glia have a capacity to regenerate neurons in lower vertebrates like zebrafish, but such ability is extremely limited in mammals. In zebrafish, Müller glia proliferate after injury, which promotes their neurogenic reprogramming while inhibiting reactive gliosis. In mammals, however, how the cell cycle affects the fate of Müller glia after injury remains unclear. Here, we focused on the expression of proneural transcription factors, Ngn2 and Ascl1, and a gliosis marker glial fibrillary acidic protein (GFAP) in rat Müller glia after N-methyl-N-nitrosourea (MNU)-induced photoreceptor injury and analyzed the role of Müller glia proliferation in the regulation of their expression using retinal explant cultures. Thymidine-induced G1/S arrest of Müller glia proliferation significantly hampered the expression of Ascl1, Ngn2, and GFAP, and release from the arrest induced their upregulation. The migration of Müller glia nuclei into the outer nuclear layer was also shown to be cell cycle-dependent. These data suggest that, unlike the situation in zebrafish, cell cycle progression of Müller glia in mammals promotes both neurogenic reprogramming and reactive gliosis, which may be one of the mechanisms underlying the limited regenerative capacity of the mammalian retina.


Gliosis , Zebrafish , Animals , Rats , Gliosis/chemically induced , Gliosis/metabolism , Transcription Factors/metabolism , Neuroglia/metabolism , Retina/metabolism , Cell Cycle , Mammals
3.
Int J Mol Sci ; 24(16)2023 Aug 12.
Article En | MEDLINE | ID: mdl-37628913

Oxidative stress with a depletion of glutathione is a key factor in the initiation and progression of Alzheimer's disease (AD). N-Acetylcysteine (NAC), a glutathione precursor, provides neuroprotective effects in AD animal models. Its amide form, N-Acetylcysteine amide (NACA), has an extended bioavailability compared to NAC. This study evaluates the neuroprotective effects of NACA against Aß1-42 peptide-induced AD-like pathology in rats. Male Wistar rats (2.5 months old) were divided into five groups: Normal Control (NC), Sham (SH), Aß, Aß + NACA and NACA + Aß + NACA (n = 8 in all groups). AD-like pathology was induced by the intracerebroventricular infusion of Aß1-42 peptide into the lateral ventricle. NACA (75 mg/kg) was administered either as a restorative (i.e., injection of NACA for 7 consecutive days after inducing AD-like pathology (Aß + N group)), or as prophylactic (for 7 days before and 7 days after inducing the pathology (N + Aß + N group)). Learning and memory, neurogenesis, expression of AD pathology markers, antioxidant parameters, neuroprotection, astrogliosis and microgliosis were studied in the hippocampus and the prefrontal cortex. All data were analyzed with a one-way ANOVA test followed by Bonferroni's multiple comparison test. NACA treatment reversed the cognitive deficits and reduced oxidative stress in the hippocampus and prefrontal cortex. Western blot analysis for Tau, Synaptophysin and Aß, as well as a histopathological evaluation through immunostaining for neurogenesis, the expression of neurofibrillary tangles, ß-amyloid peptide, synaptophysin, neuronal morphology and gliosis, showed a neuroprotective effect of NACA. In conclusion, this study demonstrates the neuroprotective effects of NACA against ß-amyloid induced AD-like pathology.


Alzheimer Disease , Neuroprotective Agents , Male , Rats , Animals , Acetylcysteine/pharmacology , Rats, Wistar , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Synaptophysin , Neuroprotective Agents/pharmacology , Amyloid beta-Peptides , Gliosis/chemically induced , Gliosis/drug therapy , Glutathione
4.
Food Chem Toxicol ; 178: 113934, 2023 Aug.
Article En | MEDLINE | ID: mdl-37423315

Evidence suggests that furan, a widespread environmental and food contaminant, causes liver toxicity and cancer, but its implications in the brain are not well defined. We measured behavioral, glial, and biochemical responses in male juvenile rats exposed orally to 2.5, 5 and 10 mg/kg furan and vitamin E after 28 days. Furan-mediated hyperactivity peaked at 5 mg/kg and did not exacerbate at 10 mg/kg. Enhanced motor defect was also observed at 10 mg/kg. Furan-treated rats elicited inquisitive exploration but showed impaired spatial working memory. Without compromising the blood-brain barrier, furan induced glial reactivity with enhanced phagocytic activity, characterized by parenchyma-wide microglial aggregation and proliferation, which switched from hyper-ramified to rod-like morphology with increasing doses. Furan altered the glutathione-S-transferase-driven enzymatic and non-enzymatic antioxidant defence systems differentially and dose-dependently across brain regions. Redox homeostasis was most perturbed in the striatum and least disrupted in hippocampus/cerebellum. Vitamin E supplementation attenuated exploratory hyperactivity and glial reactivity but did not affect impaired working memory and oxidative imbalance. Overall, sub-chronic exposure of juvenile rats to furan triggered glial reactivity and behavioral deficits suggesting the brain's vulnerability during juvenile development to furan toxicity. It remains to be determined whether environmentally relevant furan concentrations interfere with critical brain developmental milestones.


Gliosis , Neurotoxicity Syndromes , Rats , Male , Animals , Rats, Wistar , Gliosis/chemically induced , Oxidative Stress , Neurotoxicity Syndromes/etiology , Vitamin E , Furans/toxicity
5.
Brain Res ; 1815: 148444, 2023 09 15.
Article En | MEDLINE | ID: mdl-37290610

The progressive nature of acquired epilepsy warrants a thorough examination of acute changes that occur immediately after an epileptogenic insult to better understand the cellular and molecular mechanisms that trigger epileptogenesis. Astrocytes are important regulators of neuronal functions and emerging evidence suggests an involvement of astrocytic purinergic signaling in the etiology of acquired epilepsy. However, how astrocytic purinergic signaling responds immediately after an acute seizure or an epileptogenic insult to impact epileptogenesis is not well studied. In the present study, we report area-specific rapid onset of astrocytic changes in morphology, as well as in expression and functional activity of the purinergic signaling in the hippocampus that occur immediately after pilocarpine-induced stage 5 seizure. After 3 hr of stage 5 acute seizure, hippocampal astrocytes show increased intrinsic calcium activity in stratum radiatum as well as reactive astrogliosis in the stratum lacunosum moleculare and hilus regions of the hippocampus. Hilar astrocytes also upregulated the expression of P2Y1 and P2Y2 metabotropic purinergic receptors. Subsequently, P2Y1 exhibited a functional upregulation by showing a significantly higher intracellular calcium rise in ex-vivo hippocampal slices on P2Y1 activation. Our results suggest that hippocampal astrocytes undergo rapid area-specific morphological and functional changes immediately after the commencement of the seizure activity and purinergic receptors upregulation is one of the earliest changes in response to seizure activity. These changes can be considered acute astrocytic responses to seizure activity which can potentially drive the epileptogenesis and can be explored further to identify astrocyte-specific targets for seizure therapy.


Epilepsy , Pilocarpine , Rats , Animals , Calcium/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Seizures/chemically induced , Seizures/metabolism , Hippocampus/metabolism , Epilepsy/chemically induced , Epilepsy/metabolism , Astrocytes/metabolism
6.
J Pain ; 24(11): 1980-1993, 2023 11.
Article En | MEDLINE | ID: mdl-37315729

Chemotherapy-induced peripheral neuropathy (CIPN) is a common, dose-limiting side effect of cancer therapy. Protease-activated receptor 2 (PAR2) is implicated in a variety of pathologies, including CIPN. In this study, we demonstrate the role of PAR2 expressed in sensory neurons in a paclitaxel (PTX)-induced model of CIPN in mice. PAR2 knockout/wildtype (WT) mice and mice with PAR2 ablated in sensory neurons were treated with PTX administered via intraperitoneal injection. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. We then examined immunohistochemical staining of dorsal root ganglion (DRG) and hind paw skin samples from CIPN mice to measure satellite cell gliosis and intra-epidermal nerve fiber (IENF) density. The pharmacological reversal of CIPN pain was tested with the PAR2 antagonist C781. Mechanical allodynia caused by PTX treatment was alleviated in PAR2 knockout mice of both sexes. In the PAR2 sensory neuronal conditional knockout (cKO) mice, both mechanical allodynia and facial grimacing were attenuated in mice of both sexes. In the DRG of the PTX-treated PAR2 cKO mice, satellite glial cell activation was reduced compared to control mice. IENF density analysis of the skin showed that the PTX-treated control mice had a reduction in nerve fiber density while the PAR2 cKO mice had a comparable skin innervation as the vehicle-treated animals. Similar results were seen with satellite cell gliosis in the DRG, where gliosis induced by PTX was absent in PAR cKO mice. Finally, C781 was able to transiently reverse established PTX-evoked mechanical allodynia. PERSPECTIVE: Our work demonstrates that PAR2 expressed in sensory neurons plays a key role in PTX-induced mechanical allodynia, spontaneous pain, and signs of neuropathy, suggesting PAR2 as a possible therapeutic target in multiple aspects of PTX CIPN.


Paclitaxel , Peripheral Nervous System Diseases , Male , Female , Mice , Animals , Paclitaxel/adverse effects , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Receptor, PAR-2/genetics , Receptor, PAR-2/therapeutic use , Gliosis/chemically induced , Gliosis/complications , Gliosis/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Pain/complications , Sensory Receptor Cells , Mice, Knockout , Ganglia, Spinal
7.
J Neuroinflammation ; 20(1): 149, 2023 Jun 24.
Article En | MEDLINE | ID: mdl-37355700

BACKGROUND: Chemotherapy-induced neuropathic pain (CIPN) describes a pathological pain state that occurs dose-dependently as a side effect and can limit or even impede an effective cancer therapy. Unfortunately, current treatment possibilities for CIPN are remarkably confined and mostly inadequate as CIPN therapeutics themselves consist of low effectiveness and may induce severe side effects, pointing out CIPN as pathological entity with an emerging need for novel treatment targets. Here, we investigated whether the novel and highly specific FKBP51 inhibitor SAFit2 reduces paclitaxel-induced neuropathic pain. METHODS: In this study, we used a well-established multiple low-dose paclitaxel model to investigate analgesic and anti-inflammatory properties of SAFit2. For this purpose, the behavior of the mice was recorded over 14 days and the mouse tissue was then analyzed using biochemical methods. RESULTS: Here, we show that SAFit2 is capable to reduce paclitaxel-induced mechanical hypersensitivity in mice. In addition, we detected that SAFit2 shifts lipid levels in nervous tissue toward an anti-inflammatory and pro-resolving lipid profile that counteracts peripheral sensitization after paclitaxel treatment. Furthermore, SAFit2 reduced the activation of astrocytes and microglia in the spinal cord as well as the levels of pain-mediating chemokines. Its treatment also increased anti-inflammatory cytokines levels in neuronal tissues, ultimately leading to a resolution of neuroinflammation. CONCLUSIONS: In summary, SAFit2 shows antihyperalgesic properties as it ameliorates paclitaxel-induced neuropathic pain by reducing peripheral sensitization and resolving neuroinflammation. Therefore, we consider SAFit2 as a potential novel drug candidate for the treatment of paclitaxel-induced neuropathic pain.


Neuralgia , Paclitaxel , Mice , Animals , Paclitaxel/toxicity , Neuroinflammatory Diseases , Gliosis/chemically induced , Gliosis/drug therapy , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/prevention & control , Lipids/adverse effects
8.
Exp Eye Res ; 231: 109485, 2023 06.
Article En | MEDLINE | ID: mdl-37080381

The vesicant sulfur mustard (SM) is a chemical warfare agent that causes acute and chronic injury to the cornea and proximal anterior segment structures. Despite clinical evidence of SM-exposure causing unexplained retinal deficits, there have been no animal studies conducted to examine the retinal toxicity of this vesciant. The cardinal hallmark of retinal response to stressors or injury is the activation of reactive gliosis, a cellular process largely governed by Müller glia. Previously we showed that corneal exposure to sodium hydroxide elicits rapid induction of reactive gliosis and results in retinal degeneration in a dose-related manner. Based on this evidence, we hypothesized that the vesicant nitrogen mustard (NM), an analog of SM, may also elicit reactive gliosis. To test this idea, we developed a mouse model of NM ocular injury and investigated corneal and retinal effects focusing on citrullination, a posttranslational modification (PTM) of proteins. This PTM was recently linked to alkali injury and has also been shown to occur in retinal degenerative conditions. Here, we demonstrate that corneal exposure to 1% NM causes a synchronous activation of citrullination in both the cornea and retina with hypercitrullination becoming apparent temporally and manifesting with altered cellular expression characteristics. A key finding is that ocular citrullination occurs acutely as early as 1-h post-injury in both the cornea and retina, which underscores a need for expeditious interception of this acute corneal and retinal response. Moreover, exploiting dose response and temporal studies, we uncoupled NM-induced retinal citrullination from its induction of retinal gliosis. Our findings demonstrate that hypercitrullination is a common corneo-retinal mechanism that sensitizes the eye to NM injury and suggests that counteracting hypercitrullination may provide a suitable countermeasure to vesicant injury.


Eye Injuries , Mustard Gas , Retinal Diseases , Animals , Mice , Mechlorethamine/toxicity , Irritants/adverse effects , Irritants/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Cornea/metabolism , Eye Injuries/chemically induced , Eye Injuries/metabolism , Retina , Mustard Gas/toxicity , Retinal Diseases/chemically induced , Retinal Diseases/metabolism
9.
Chem Biol Interact ; 375: 110428, 2023 Apr 25.
Article En | MEDLINE | ID: mdl-36868496

Cadmium (Cd) produces cognition decline following single and repeated treatment, although the complete mechanisms are still unrevealed. Basal forebrain (BF) cholinergic neurons innervate the cortex and hippocampus, regulating cognition. Cd single and repeated exposure induced BF cholinergic neuronal loss, partly through thyroid hormones (THs) disruption, which may cause the cognition decline observed following Cd exposure. However, the mechanisms through which THs disruption mediate this effect remain unknown. To research the possible mechanisms through which Cd-induced THs deficiency may mediate BF neurodegeneration, Wistar male rats were treated with Cd for 1- (1 mg/kg) or 28-days (0.1 mg/kg) with or without triiodothyronine (T3, 40 µg/kg/day). Cd exposure promoted neurodegeneration, spongiosis, gliosis and several mechanisms related to these alterations (increased H202, malondialdehyde, TNF-α, IL-1ß, IL-6, BACE1, Aß and phosphorylated-Tau levels, and decreased phosphorylated-AKT and phosphorylated-GSK-3ß levels). T3 supplementation partially reversed the effects observed. Our results show that Cd induces several mechanisms that may be responsible for the neurodegeneration, spongiosis and gliosis observed in the rats' BF, which are partially mediated by a reduction in THs levels. These data may help to explain the mechanisms through which Cd induces BF neurodegeneration, possibly leading to the cognitive decline observed, providing new therapeutic tools to prevent and treat these damages.


Basal Forebrain , Cadmium , Animals , Male , Rats , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Basal Forebrain/metabolism , Cadmium/toxicity , Gliosis/chemically induced , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation , Rats, Wistar , Reactive Oxygen Species , tau Proteins/metabolism , Thyroid Hormones
10.
J Neurosci Res ; 101(6): 916-929, 2023 06.
Article En | MEDLINE | ID: mdl-36696411

Deep brain stimulation (DBS) of the anterior nucleus of the thalamus (ANT) appears to be effective against seizures in animals and humans however, its therapeutic mechanisms remain elusive. This study aimed to combine 9.4T multimodal magnetic resonance imaging (MRI) with histology to investigate the longitudinal effects of long-term ANT-DBS in pilocarpine-induced epileptic rats. Status epilepsy (SE) was induced by LiCl-pilocarpine injection in 11 adult male Sprague-Dawley rats. Four weeks after SE, chronic epileptic rats underwent either ANT-DBS (n = 6) or sham-DBS (n = 5) surgery. Electroencephalography (EEG) and spontaneous recurrent seizures (SRS) were recorded for 1 week. The T2-weighted image and images from resting-state functional MRI (rs-fMRI) were acquired at three states: before SE, at 4 weeks post-SE, and at 5 weeks post-DBS. Volumes of the hippocampal subregions and hippocampal-related functional connectivity (FC) were compared longitudinally. Finally, antibodies against neuronal nuclei (NeuN) and glial fibrillary acidic proteins were used to evaluate neuronal loss and astrogliosis in the hippocampus. Long-term ANT-DBS significantly reduced seizure generalization in pilocarpine-induced epileptic rats. By analyzing the gray matter volume using T2-weighted images, long-term ANT-DBS displayed morphometric restoration of the hippocampal subregions. Neuronal protection of the hippocampal subregions and inhibition of astrogliosis in the hippocampal subregions were observed in the ANT-DBS group. ANT-DBS caused reversible regulation of FC in the insula-hippocampus and subthalamic nucleus-hippocampus. Long-term ANT-DBS provides comprehensive protection of hippocampal histology, hippocampal morphometrics, and hippocampal-related functional networks.


Deep Brain Stimulation , Epilepsy , Humans , Adult , Rats , Male , Animals , Pilocarpine/toxicity , Pilocarpine/metabolism , Gliosis/chemically induced , Gliosis/diagnostic imaging , Gliosis/metabolism , Rats, Sprague-Dawley , Deep Brain Stimulation/methods , Epilepsy/chemically induced , Epilepsy/diagnostic imaging , Epilepsy/therapy , Seizures/metabolism , Magnetic Resonance Imaging , Hippocampus/metabolism
11.
Brain Res ; 1801: 148202, 2023 02 15.
Article En | MEDLINE | ID: mdl-36521513

Obstructive sleep apnea is highly prevalent in Alzheimer's disease (AD). However, brainstem centers controlling respiration have received little attention in AD research, and mechanisms behind respiratory dysfunction in AD are not understood. The nucleus tractus solitarii (nTS) is an important brainstem center for respiratory control and chemoreflex function. Alterations of nTS integrity, like those shown in AD patients, likely affect neuronal processing and adequate control of breathing. We used the streptozotocin-induced rat model of AD (STZ-AD) to analyze cellular changes in the nTS that corroborate previously documented respiratory dysfunction. We used 2 common dosages of STZ (2 and 3 mg/kg STZ) for model induction and evaluated the early impact on cell populations in the nTS. The hippocampus served as control region to identify site-specific effects of STZ. There was significant atrophy in the caudal nTS of the 3 mg/kg STZ-AD group only, an area known to integrate chemoafferent information. Also, the hippocampus had significant atrophy with the highest STZ dosage tested. Both STZ-AD groups showed respiratory dysfunction along with multiple indices for astroglial and microglial activation. These changes were primarily located in the caudal and intermediate nTS. While there was no change of astrocytes in the hippocampus, microglial activation was accompanied by a reduction in synaptic density. Together, our data demonstrate that STZ-AD induces site-specific effects on all major cell types, primarily in the caudal/intermediate nTS. Both STZ dosages used in this study produced a similar outcome and can be used for future studies examining the initial symptoms of STZ-AD.


Alzheimer Disease , Solitary Nucleus , Rats , Animals , Solitary Nucleus/metabolism , Streptozocin/pharmacology , Gliosis/chemically induced , Gliosis/metabolism , Alzheimer Disease/metabolism , Respiration
12.
Neurotoxicology ; 94: 59-70, 2023 01.
Article En | MEDLINE | ID: mdl-36336098

Pyrrolizidine alkaloids (PAs) are secondary plant metabolites playing an important role as phytotoxins in the plant defense mechanisms and can be present as contaminant in the food of humans and animals. The PA monocrotaline (MCT), one of the major plant derived toxin that affect humans and animals, is present in a high concentration in Crotalaria spp. (Leguminosae) seeds and can induce toxicity after consumption, characterized mainly by hepatotoxicity and pneumotoxicity. However, the effects of the ingestion of MCT in the central nervous system (CNS) are still poorly elucidated. Here we investigated the effects of MCT oral acute administration on the behavior and CNS toxicity in rats. Male adult Wistar were treated with MCT (109 mg/Kg, oral gavage) and three days later the Elevated Pluz Maze test demonstrated that MCT induced an anxiolytic-like effect, without changes in novelty habituation and in operational and spatial memory profiles. Histopathology revealed that the brain of MCT-intoxicated animals presented hyperemic vascular structures in the hippocampus, parahippocampal cortex and neocortex, mild perivascular edema in the neocortex, hemorrhagic focal area in the brain stem, hemorrhage and edema in the thalamus. MCT also induced neurotoxicity in the cortex and hippocampus, as revealed by Fluoro Jade-B and Cresyl Violet staining, as well astrocyte reactivity, revealed by immunocytochemistry for glial fibrillary acidic protein. Additionally, it was demonstrated by RT-qPCR that MCT induced up-regulation on mRNA expression of neuroinflammatory mediator, especially IL1ß and CCL2 in the hippocampus and cortex, and down-regulation on mRNA expression of neurotrophins HGDF and BDNF in the cortex. Together, these results demonstrate that the ingestion of MCT induces cerebrovascular lesions and toxicity to neurons that are associated to astroglial cell response and neuroinflammation in the cortex and hippocampus of rats, highlighting CNS damages after acute intoxication, also putting in perspective it uses as a model for cerebrovascular damage.


Gliosis , Monocrotaline , Humans , Rats , Animals , Monocrotaline/toxicity , Monocrotaline/metabolism , Gliosis/chemically induced , Rats, Wistar , Astrocytes/metabolism , RNA, Messenger/metabolism
13.
Neurochem Res ; 48(5): 1468-1479, 2023 May.
Article En | MEDLINE | ID: mdl-36502418

The reactivation of astrocytes plays a critical role in spinal cord injury (SCI) repairment. In this study, IL1RAP expression has been found to be upregulated in SCI mice spinal cord, SCI astrocytes, and LPS-stimulated NHAs. Genes correlated with IL1RAP were significantly enriched in cell proliferation relative pathways. In LPS-stimulated NHAs, IL1RAP overexpression promoted NHA cell proliferation, decreased PTEN protein levels, and increased the phosphorylation of Akt and mTOR. IL1RAP overexpression promoted LPS-induced NHA activation and NF-κB signaling activation. Conditioned medium from IL1RAP-overexpressing NHAs inhibited SH-SY5Y cells viability but promoted cell apoptosis. Conclusively, IL1RAP knockdown in LPS-stimulated NHAs could partially suppress LPS-induced reactive astrogliosis, therefore promoting neuronal cell proliferation.


Neuroblastoma , Spinal Cord Injuries , Humans , Mice , Animals , Lipopolysaccharides/toxicity , Astrocytes/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Neuroblastoma/metabolism , Cell Proliferation/physiology , Spinal Cord Injuries/metabolism , Interleukin-1 Receptor Accessory Protein/metabolism
14.
Neurotoxicology ; 88: 57-64, 2022 01.
Article En | MEDLINE | ID: mdl-34728274

High ethanol (EtOH) consumption is a serious condition that induces tremors, alcoholic psychosis, and delirium, being considered a public health problem worldwide. Prolonged EtOH exposure promotes neurodegeneration, affecting several neurotransmitter systems and transduction signaling pathways. Glutamate is the major excitatory amino acid in the central nervous system (CNS) and the extracellular glutamatergic tonus is controlled by glutamate transporters mostly located in astrocytes. Here, we explore the effects of prolonged EtOH exposure on the glutamatergic uptake system and its relationship with astroglial markers (GFAP and S100B), neuroinflammation (IL-1ß and TNF-α), and brain derived neurotrophic factor (BDNF) levels in the CNS of adult zebrafish. Animals were exposed to 0.5% EtOH for 7, 14, and 28 days continuously. Glutamate uptake was significantly decreased after 7 and 14 days of EtOH exposure, returning to baseline levels after 28 days of exposure. No alterations were observed in crucial enzymatic activities linked to glutamate uptake, like Na,K-ATPase or glutamine synthetase. Prolonged EtOH exposure increased GFAP, S100B, and TNF-α levels after 14 days. Additionally, increased BDNF mRNA levels were observed after 14 and 28 days of EtOH exposure, while BDNF protein levels increased only after 28 days. Collectively, our data show markedly brain astroglial, neuroinflammatory and neurotrofic responses after an initial impairment of glutamate uptake following prolonged EtOH exposure. This neuroplasticity event could play a key role in the modulatory effect of EtOH on glutamate uptake after 28 days of continuous exposure.


Brain/drug effects , Ethanol/adverse effects , Gliosis/chemically induced , Glutamic Acid/metabolism , Neuroinflammatory Diseases/chemically induced , Animals , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Female , Gliosis/pathology , Interleukin-1beta/metabolism , Male , Neuroinflammatory Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Potassium-Exchanging ATPase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Zebrafish , Zebrafish Proteins/metabolism
15.
Alcohol Clin Exp Res ; 46(2): 207-220, 2022 02.
Article En | MEDLINE | ID: mdl-34862633

BACKGROUND: Multiple ethanol binge drinking-like exposures during adolescence in the rat induce neuroinflammation, loss of neurogenesis, and cognitive deficits in adulthood. Interestingly, the first ethanol binge drinking-like exposure during adolescence also induces short- term impairments in cognition and synaptic plasticity in the hippocampus though the cellular mechanisms of these effects are unclear. Here, we sought to determine which of the cellular effects of ethanol might play a role in the disturbances in cognition and synaptic plasticity observed in the adolescent male rat after two binge-like ethanol exposures. METHODS: Using immunochemistry, we measured neurogenesis, neuronal loss, astrogliosis, neuroinflammation, and synaptogenesis in the hippocampus of adolescent rats 48 h after two binge-like ethanol exposures (3 g/kg, i.p., 9 h apart). We used flow cytometry to analyze activated microglia and identify the TLR4-expressing cell types. RESULTS: We detected increased hippocampal doublecortin immunoreactivity in the subgranular zone (SGZ) of the dentate gyrus (DG), astrogliosis in the SGZ, and a reduced number of mature neurons in the DG and in CA3, suggesting compensatory neurogenesis. Synaptic density decreased in the stratum oriens of CA1 revealing structural plasticity. There was no change in microglial TLR4 expression or in the number of activated microglia, suggesting a lack of neuroinflammatory processes, although neuronal TLR4 was decreased in CA1 and DG. CONCLUSIONS: Our findings demonstrate that the cognitive deficits associated with hippocampal synaptic plasticity alterations that we previously characterized 48 h after the first binge-like ethanol exposures are associated with hippocampal structural plasticity, astrogliosis, and decreased neuronal TLR4 expression, but not with microglia reactivity.


Binge Drinking/physiopathology , Ethanol/pharmacology , Gliosis/chemically induced , Neurogenesis/drug effects , Animals , Binge Drinking/complications , Cognitive Dysfunction/chemically induced , Ethanol/administration & dosage , Hippocampus/drug effects , Male , Microglia/metabolism , Neuronal Plasticity/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley
16.
Chem Biol Interact ; 351: 109740, 2022 Jan 05.
Article En | MEDLINE | ID: mdl-34742682

Lead (Pb) exposure can cause damage to the central nervous system (CNS)*. Pb can accumulate in the hippocampus, leading to learning and memory impairments. Recent studies have shown that high-fat diet (HFD) is also associated with cognitive impairment. However, there are few reports on CNS damage due to HFD and Pb exposure. We aimed to investigate the effect of Pb on cognitive functions of HFD-fed mice, focusing on the role of regulatory T (Treg) cells in astrocyte activation. C57BL/6J mice were randomly divided into control, HFD, Pb, and HFD + Pb groups. TGF-ß and IL-10 secreted by Treg cells and the intracellular transcription factor Foxp3 were evaluated as a measure of Treg cell function; astrocyte activation was assessed by evaluating glial fibrillary acidic protein (GFAP) expression. The learning and memory ability was significantly lower in the HFD + Pb group than in other groups. The brain Treg cell ratio was significantly decreased and the protein levels of TGF-ß, IL-10, and Foxp3 were significantly lower, whereas the protein level of GFAP was higher in the HFD + Pb group. The hippocampus of the HFD + Pb group mice showed significantly higher levels of neurotoxic reactive astrocyte markers and astrogliosis was also much higher compared to HFD and Pb groups. Furthermore, all-trans retinoic acid treatment increased the brain Treg cell ratio, reversed cognitive decline, and suppressed astrocyte activation in the HFD + Pb group mice. We concluded that HFD along with Pb exposure could aggravate the activation of astrocytes in the brain, and the brain Treg cells may be involved in inhibiting astrocyte activation in HFD-fed mice exposed to Pb.


Astrocytes/metabolism , Brain/metabolism , Cognitive Dysfunction/metabolism , Diet, High-Fat/adverse effects , Lead/toxicity , T-Lymphocytes, Regulatory/metabolism , Animals , Astrocytes/drug effects , Brain/drug effects , Cognitive Dysfunction/chemically induced , Gliosis/chemically induced , Learning/drug effects , Male , Memory/drug effects , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects
17.
Sci Rep ; 11(1): 23404, 2021 12 03.
Article En | MEDLINE | ID: mdl-34862457

Prenatal inflammation negatively affects placental function, subsequently altering fetal development. Pathogen-associated molecular patterns (PAMPs) are used to mimics infections in preclinical models but rarely detected during pregnancy. Our group previously developed an animal model of prenatal exposure to uric acid (endogenous mediator), leading to growth restriction alongside IL-1-driven placental inflammation (Brien et al. in J Immunol 198(1):443-451, 2017). Unlike PAMPs, the postnatal impact of prenatal non-pathogenic inflammation is still poorly understood. Therefore, we investigated the effects of prenatal uric acid exposure on postnatal neurodevelopment and the therapeutic potential of the IL-1 receptor antagonist; IL-1Ra. Uric acid induced growth restriction and placental inflammation, which IL-1Ra protected against. Postnatal evaluation of both structural and functional aspects of the brain revealed developmental changes. Both astrogliosis and microgliosis were observed in the hippocampus and white matter at postnatal day (PND)7 with IL-1Ra being protective. Decreased myelin density was observed at PND21, and reduced amount of neuronal precursor cells was observed in the Dentate Gyrus at PND35. Functionally, motor impairments were observed as evaluated with the increased time to fully turn upward (180 degrees) on the inclined plane and the pups were weaker on the grip strength test. Prenatal exposure to sterile inflammation, mimicking most clinical situation, induced growth restriction with negative impact on neurodevelopment. Targeted anti-inflammatory intervention prenatally could offer a strategy to protect brain development during pregnancy.


Fetal Growth Retardation/drug therapy , Gliosis/drug therapy , Inflammation/drug therapy , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Placenta/drug effects , Uric Acid/adverse effects , Animals , Animals, Newborn , Brain/drug effects , Brain/embryology , Disease Models, Animal , Female , Fetal Development/drug effects , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/immunology , Gliosis/chemically induced , Gliosis/immunology , Inflammation/chemically induced , Interleukin 1 Receptor Antagonist Protein/pharmacology , Placenta/immunology , Pregnancy , Rats , Rats, Sprague-Dawley
18.
Int J Mol Sci ; 22(21)2021 Oct 20.
Article En | MEDLINE | ID: mdl-34768765

The exact etiology of Parkinson's disease (PD) remains largely unknown, but more and more research suggests the involvement of the gut microbiota. Interestingly, idiopathic PD patients were shown to have at least a 10 times higher prevalence of Helicobacter suis (H. suis) DNA in gastric biopsies compared to control patients. H. suis is a zoonotic Helicobacter species that naturally colonizes the stomach of pigs and non-human primates but can be transmitted to humans. Here, we investigated the influence of a gastric H. suis infection on PD disease progression through a 6-hydroxydopamine (6-OHDA) mouse model. Therefore, mice with either a short- or long-term H. suis infection were stereotactically injected with 6-OHDA in the left striatum and sampled one week later. Remarkably, a reduced loss of dopaminergic neurons was seen in the H. suis/6-OHDA groups compared to the control/6-OHDA groups. Correspondingly, motor function of the H. suis-infected 6-OHDA mice was superior to that in the non-infected 6-OHDA mice. Interestingly, we also observed higher expression levels of antioxidant genes in brain tissue from H. suis-infected 6-OHDA mice, as a potential explanation for the reduced 6-OHDA-induced cell loss. Our data support an unexpected neuroprotective effect of gastric H. suis on PD pathology, mediated through changes in oxidative stress.


Helicobacter Infections , Helicobacter heilmannii/physiology , Parkinson Disease/microbiology , Stomach/microbiology , Animals , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/microbiology , Female , Gliosis/chemically induced , Gliosis/microbiology , Helicobacter heilmannii/growth & development , Inflammation/microbiology , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents , Oxidative Stress/physiology , Oxidopamine/toxicity , Parkinson Disease/complications , Parkinson Disease/pathology , Peroxidases/genetics , Peroxidases/metabolism , Stomach Diseases/physiopathology
19.
J Neurochem ; 159(6): 1016-1027, 2021 12.
Article En | MEDLINE | ID: mdl-34699606

Reactive astrogliosis is an early event in Alzheimer's disease (AD) brain and plays a key role in synaptic degeneration in AD development. Zinc accumulates in extracellular fraction and synaptosomes in AD human brains with its effect on reactive astrocytes remaining unknown. Through Western blotting, Quantitative polymerase chain reaction (qPCR), and immunofluorescence detection on primary astrocytes treated by zinc and/or zinc chelator, we revealed that zinc induced harmful A1-type reactive astrogliosis in cultured primary astrocytes; the latter, promoted synaptic degeneration in primary neurons. The mechanism investigation showed that zinc induced activation of extracellular regulated protein kinase (ERK) and Janus kinase 2 (JAK2), which phosphorylated signal transduction and transcription activator 3 (Stat3) at serine 727 (S727-Stat3) and tyrosine 705 (Y705-Stat3), respectively, resulting in activation of Stat3. Stat3 phosphorylation at S727 by ERK plays a key role in zinc-induced astrogliosis. These data imply a new molecular mechanism of reactive astrogliosis in AD, in which excessive zinc activates Stat3 through up-regulating ERK signaling pathway.


Gliosis/metabolism , MAP Kinase Signaling System/physiology , Nerve Degeneration/metabolism , STAT3 Transcription Factor/metabolism , Synapses/metabolism , Zinc/toxicity , Animals , Animals, Newborn , Cells, Cultured , Female , Gliosis/chemically induced , Gliosis/pathology , MAP Kinase Signaling System/drug effects , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Pregnancy , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/pathology
20.
J Chem Neuroanat ; 117: 102011, 2021 11.
Article En | MEDLINE | ID: mdl-34384873

Autism spectrum disorder (ASD) is a range of neurodevelopmental disorders characterized by movement and social deficits with rapidly increasing incidence worldwide. Propionic acid (PPA) is a histone deacetylase inhibitor that regulates neuronal plasticity in the brain. Evaluation of the behavioral and cellular consequences of PPA exposure during a critical neurodevelopmental window is required. Therefore, in the present study we aimed to evaluate the effects of prenatal PPA exposure on locomotor behavior and astrocyte number, as well as on levels of nitric oxide (NO), synaptophysin (SYP; a marker of synaptic plasticity), and metallothionein 3 (MT-III; a marker of reactive oxygen species and zinc metabolism), in the prefrontal cortex (PFC) of male rats. All parameters were evaluated at three critical ages of development: postnatal days (PD) 21 (weaning age), PD35 (pre-pubertal age) and PD70 (post-pubertal age). Prenatal PPA exposure induced hypolocomotion and decreased rearing events at weaning age. Moreover, astrogliosis in the PFC was observed in PPA-treated rats at pre- and post-pubertal age. SYP levels were dramatically decreased in PPA-treated rats with simultaneous astrogliosis, suggesting reduced synaptic plasticity. MT-III expression was deregulated in PPA-treated rats. Finally, the expression of NO in the PFC remained unaltered in PPA-treated rats. These results mimic behavioral, neuronal and astrocytic characteristics observed in ASD patients.


Gliosis/chemically induced , Gliosis/pathology , Locomotion/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Propionates/toxicity , Age Factors , Animals , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/pathology , Female , Locomotion/physiology , Male , Pregnancy , Rats , Rats, Sprague-Dawley
...