Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Neuroscience ; 549: 65-75, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38750924

ABSTRACT

Recent evidence has supported a pathogenic role for neuroinflammation in Parkinson's disease (PD). Inflammatory response has been associated with symptoms and subtypes of PD. However, it is unclear whether immune changes are involved in the initial pathogenesis of PD, leading to the non-motor symptoms (NMS) observed in its prodromal stage. The current study aimed to characterize the behavioral and cognitive changes in a toxin-induced model of prodromal PD-like syndrome. We also sought to investigate the role of neuroinflammation in prodromal PD-related NMS. Male mice were subjected to bilateral intranasal infusion with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or saline (control group), followed by comprehensive behavioral, pathological and neurochemical analysis. Intranasal MPTP infusion was able to cause the loss of dopaminergic neurons in the substantia nigra (SN). In parallel, it induced impairment in olfactory discrimination and social memory consolidation, compulsive and anxiety-like behaviors, but did not influence motor performance. Iba-1 and GFAP expressions were increased in the SN, suggesting an activated state of microglia and astrocytes. Consistent with this, MPTP mice had increased levels of IL-10 and IL-17A, and decreased levels of BDNF and TrkA mRNA in the SN. The striatum showed increased IL-17A, BDNF, and NFG levels compared to control mice. In conclusion, neuroinflammation may play an important role in the early stage of experimental PD-like syndrome, leading to cognitive and behavioral changes. Our results also indicate that intranasal administration of MPTP may represent a valuable mouse model for prodromal PD.


Subject(s)
Disease Models, Animal , Mice, Inbred C57BL , Prodromal Symptoms , Substantia Nigra , Animals , Male , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/drug effects , Dopaminergic Neurons/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Neuroinflammatory Diseases/pathology , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Mice , Microglia/metabolism , Microglia/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Brain-Derived Neurotrophic Factor/metabolism , Anxiety/etiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology
2.
Biomolecules ; 14(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38672445

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal motoneuron degenerative disease that is associated with demyelination. The Wobbler (WR) mouse exhibits motoneuron degeneration, gliosis and myelin deterioration in the cervical spinal cord. Since male WRs display low testosterone (T) levels in the nervous system, we investigated if T modified myelin-relative parameters in WRs in the absence or presence of the aromatase inhibitor, anastrozole (A). We studied myelin by using luxol-fast-blue (LFB) staining, semithin sections, electron microscopy and myelin protein expression, density of IBA1+ microglia and mRNA expression of inflammatory factors, and the glutamatergic parameters glutamine synthetase (GS) and the transporter GLT1. Controls and WR + T showed higher LFB, MBP and PLP staining, lower g-ratios and compact myelin than WRs and WR + T + A, and groups showing the rupture of myelin lamellae. WRs showed increased IBA1+ cells and mRNA for CD11b and inflammatory factors (IL-18, TLR4, TNFαR1 and P2Y12R) vs. controls or WR + T. IBA1+ cells, and CD11b were not reduced in WR + T + A, but inflammatory factors' mRNA remained low. A reduction of GS+ cells and GLT-1 immunoreactivity was observed in WRs and WR + T + A vs. controls and WR + T. Clinically, WR + T but not WR + T + A showed enhanced muscle mass, grip strength and reduced paw abnormalities. Therefore, T effects involve myelin protection, a finding of potential clinical translation.


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Myelin Sheath , Testosterone , Animals , Mice , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Male , Testosterone/pharmacology , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/genetics , Microglia/drug effects , Microglia/metabolism , Microglia/pathology
3.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673763

ABSTRACT

Chronic ethanol exposure often triggers neuroinflammation in the brain's reward system, potentially promoting the drive for ethanol consumption. A main marker of neuroinflammation is the microglia-derived monocyte chemoattractant protein 1 (MCP1) in animal models of alcohol use disorder in which ethanol is forcefully given. However, there are conflicting findings on whether MCP1 is elevated when ethanol is taken voluntarily, which challenges its key role in promoting motivation for ethanol consumption. Here, we studied MCP1 mRNA levels in areas implicated in consumption motivation-specifically, the prefrontal cortex, hippocampus, and striatum-as well as in the cerebellum, a brain area highly sensitive to ethanol, of C57BL/6 mice subjected to intermittent and voluntary ethanol consumption for two months. We found a significant increase in MCP1 mRNA levels in the cerebellum of mice that consumed ethanol compared to controls, whereas no significant changes were observed in the prefrontal cortex, hippocampus, or striatum or in microglia isolated from the hippocampus and striatum. To further characterize cerebellar neuroinflammation, we measured the expression changes in other proinflammatory markers and chemokines, revealing a significant increase in the proinflammatory microRNA miR-155. Notably, other classical proinflammatory markers, such as TNFα, IL6, and IL-1ß, remained unaltered, suggesting mild neuroinflammation. These results suggest that the onset of neuroinflammation in motivation-related areas is not required for high voluntary consumption in C57BL/6 mice. In addition, cerebellar susceptibility to neuroinflammation may be a trigger to the cerebellar degeneration that occurs after chronic ethanol consumption in humans.


Subject(s)
Alcohol Drinking , Cerebellum , Chemokine CCL2 , Corpus Striatum , Ethanol , Hippocampus , Mice, Inbred C57BL , Prefrontal Cortex , Animals , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Mice , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Cerebellum/metabolism , Cerebellum/drug effects , Cerebellum/pathology , Male , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/drug effects , Ethanol/adverse effects , Alcohol Drinking/adverse effects , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/pathology , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/chemically induced
4.
J Neurosci Res ; 102(4): e25336, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38656664

ABSTRACT

Chronic neuroinflammation has been implicated in neurodegenerative disease pathogenesis. A key feature of neuroinflammation is neuronal loss and glial activation, including microglia and astrocytes. 4R-cembranoid (4R) is a natural compound that inhibits hippocampal pro-inflammatory cytokines and increases memory function in mice. We used the lipopolysaccharide (LPS) injection model to study the effect of 4R on neuronal density and microglia and astrocyte activation. C57BL/6J wild-type mice were injected with LPS (5 mg/kg) and 2 h later received either 4R (6 mg/kg) or vehicle. Mice were sacrificed after 72 h for analysis of brain pathology. Confocal images of brain sections immunostained for microglial, astrocyte, and neuronal markers were used to quantify cellular hippocampal phenotypes and neurons. Hippocampal lysates were used to measure the expression levels of neuronal nuclear protein (NeuN), inducible nitrous oxide synthase (iNOS), arginase-1, thrombospondin-1 (THBS1), glial cell-derived neurotrophic factor (GDNF), and orosomucoid-2 (ORM2) by western blot. iNOS and arginase-1 are widely used protein markers of pro- and anti-inflammatory microglia, respectively. GDNF promotes neuronal survival, and ORM2 and THBS1 are astrocytic proteins that regulate synaptic plasticity and inhibit microglial activation. 4R administration significantly reduced neuronal loss and the number of pro-inflammatory microglia 72 h after LPS injection. It also decreased the expression of the pro-inflammatory protein iNOS while increasing arginase-1 expression, supporting its anti-inflammatory role. The protein expression of THBS1, GDNF, and ORM2 was increased by 4R. Our data show that 4R preserves the integrity of hippocampal neurons against LPS-induced neuroinflammation in mice.


Subject(s)
Hippocampus , Lipopolysaccharides , Mice, Inbred C57BL , Neuroglia , Neurons , Animals , Lipopolysaccharides/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/drug therapy , Phenotype , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology
5.
Curr Cancer Drug Targets ; 24(6): 579-594, 2024.
Article in English | MEDLINE | ID: mdl-38310461

ABSTRACT

Glioblastoma (GBM) stands as the most aggressive and lethal among the main types of primary brain tumors. It exhibits malignant growth, infiltrating the brain tissue, and displaying resistance toward treatment. GBM is a complex disease characterized by high degrees of heterogeneity. During tumour growth, microglia and astrocytes, among other cells, infiltrate the tumour microenvironment and contribute extensively to gliomagenesis. Tumour-associated macrophages (TAMs), either of peripheral origin or representing brain-intrinsic microglia, are the most numerous nonneoplastic populations in the tumour microenvironment in GBM. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumour microenvironment, which mostly induces tumour aggressiveness and drug resistance. The immunosuppressive tumour microenvironment of GBM provides multiple pathways for tumour immune evasion, contributing to tumour progression. Additionally, TAMs and astrocytes can contribute to tumour progression through the release of cytokines and activation of signalling pathways. In this review, we summarize the role of the microenvironment in GBM progression, focusing on neuroinflammation. These recent advancements in research of the microenvironment hold the potential to offer a promising approach to the treatment of GBM in the coming times.


Subject(s)
Brain Neoplasms , Disease Progression , Glioblastoma , Neuroinflammatory Diseases , Tumor Microenvironment , Humans , Glioblastoma/pathology , Glioblastoma/immunology , Glioblastoma/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/immunology , Astrocytes/pathology , Astrocytes/metabolism , Astrocytes/immunology , Animals , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Signal Transduction , Microglia/pathology , Microglia/immunology
6.
Mol Neurobiol ; 61(8): 6119-6134, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38277116

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that involves functional and structural defects in selective central nervous system (CNS) regions, harming the individual capability to process and respond to external stimuli, including impaired verbal and non-verbal communications. Etiological causes of ASD have not been fully clarified; however, prenatal activation of the innate immune system by external stimuli might infiltrate peripheral immune cells into the fetal CNS and activate cytokine secretion by microglia and astrocytes. For instance, genomic and postmortem histological analysis has identified proinflammatory gene signatures, microglia-related expressed genes, and neuroinflammatory markers in the brain during ASD diagnosis. Active neuroinflammation might also occur during the developmental stage, promoting the establishment of a defective brain connectome and increasing susceptibility to ASD after birth. While still under investigation, we tested the hypothesis whether the monocyte chemoattractant protein-1 (MCP-1) signaling is prenatally programmed to favor peripheral immune cell infiltration and activate microglia into the fetal CNS, setting susceptibility to autism-like behavior. In this review, we will comprehensively provide the current understanding of the prenatal activation of MCP-1 signaling by external stimuli during the developmental stage as a new selective node to promote neuroinflammation, brain structural alterations, and behavioral defects associated to ASD diagnosis.


Subject(s)
Chemokine CCL2 , Signal Transduction , Humans , Chemokine CCL2/metabolism , Animals , Disease Susceptibility , Autistic Disorder/metabolism , Autistic Disorder/pathology , Female , Microglia/metabolism , Microglia/pathology , Pregnancy , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Brain/metabolism , Brain/pathology , Fetal Development/physiology
7.
Cell Mol Neurobiol ; 43(6): 2473-2490, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36935429

ABSTRACT

Microglia, the resident macrophages of the central nervous system, are essential players during physiological and pathological processes. Although they participate in synaptic pruning and maintenance of neuronal circuits, microglia are mainly studied by their activity modulating inflammatory environment and adapting their phenotype and mechanisms to insults detected in the brain parenchyma. Changes in microglial phenotypes are reflected in their morphology, membrane markers, and secreted substances, stimulating neighbor glia and leading their responses to control stimuli. Understanding how microglia react in various microenvironments, such as chronic inflammation, made it possible to establish therapeutic windows and identify synergic interactions with acute damage events like stroke. Obesity is a low-grade chronic inflammatory state that gradually affects the central nervous system, promoting neuroinflammation development. Obese patients have the worst prognosis when they suffer a cerebral infarction due to basal neuroinflammation, then obesity-induced neuroinflammation could promote the priming of microglial cells and favor its neurotoxic response, potentially worsening patients' prognosis. This review discusses the main microglia findings in the obesity context during the course and resolution of cerebral infarction, involving the temporality of the phenotype changes and balance of pro- and anti-inflammatory responses, which is lost in the swollen brain of an obese subject. Obesity enhances proinflammatory responses during a stroke. Obesity-induced systemic inflammation promotes microglial M1 polarization and priming, which enhances stroke-associated damage, increasing M1 and decreasing M2 responses.


Subject(s)
Microglia , Stroke , Humans , Microglia/pathology , Neuroinflammatory Diseases , Stroke/complications , Stroke/pathology , Inflammation/pathology , Cerebral Infarction/pathology , Obesity/complications
8.
Pharmacol Rep ; 74(5): 832-846, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36042131

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative condition characterized by cognitive and functional impairments. The investigation of AD has focused on the formation of senile plaques, composed mainly by amyloid ß (Aß) peptide, and neurofibrillary tangles (NFTs) in the brain. Senile plaques and NFTs cause the excessive recruitment and activation of microglia, thus generating neuroinflammation and neuronal damage. Among the risk factors for the development of AD, diabetes has increasingly attracted attention. Hyperglycemia, the fundamental characteristic of diabetes, is involved in several mechanisms that give rise to microglial overactivation, resulting in neuronal damage and cognitive impairment. Indeed, various studies have identified the correlation between diabetes and AD. The aim of this review is to describe various mechanisms of the hyperglycemia-induced overactivation of microglia, which leads to neuroinflammation and neuronal damage and consequently contributes to the pathology of AD. The disruption of the regulation of microglial activity by hyperglycemia occurs through many mechanisms, including a greater production of reactive oxygen species (ROS) and glycation end products (AGEs), and a decrease in the elimination of Aß. The future direction of research on the relation between hyperglycemia and AD is addressed, such as the importance of determining whether the hyperglycemia-induced harmful effects on microglial activity can be reversed or attenuated if blood glucose returns to a normal level.


Subject(s)
Alzheimer Disease , Hyperglycemia , Humans , Alzheimer Disease/pathology , Microglia/pathology , Amyloid beta-Peptides , Plaque, Amyloid/complications , Plaque, Amyloid/pathology , Reactive Oxygen Species , Blood Glucose , Hyperglycemia/complications
9.
Glycoconj J ; 39(5): 685-699, 2022 10.
Article in English | MEDLINE | ID: mdl-35653015

ABSTRACT

Neurodegeneration is a pathological condition that is associated with the loss of neuronal function and structure. In neurodegenerative diseases, mounting evidence indicates that neuroinflammation is a common factor that contributes to neuronal damage and neurodegeneration. Neuroinflammation is characterized by the activation of microglia, the neuroimmune cells of the central nervous system (CNS), which have been implicated as active contributors to neuronal damage. Glycan structure modification is defining the outcome of neuroinflammation and neuronal regeneration; moreover, the expression of galectins, a group of lectins that specifically recognize ß-galactosides, has been proposed as a key factor in neuronal regeneration and modulation of the inflammatory response. Of the different galectins identified, galectin-1 stimulates the secretion of neurotrophic factors in astrocytes and promotes neuronal regeneration, whereas galectin-3 induces the proliferation of microglial cells and modulates cell apoptosis. Galectin-8 emerged as a neuroprotective factor, which, in addition to its immunosuppressive function, could generate a neuroprotective environment in the brain. This review describes the role of galectins in the activation and modulation of astrocytes and microglia and their anti- and proinflammatory functions within the context of neuroinflammation. Furthermore, it discusses the potential use of galectins as a therapeutic target for the inflammatory response and remodeling in damaged tissues in the central nervous system.


Subject(s)
Neurodegenerative Diseases , Astrocytes/metabolism , Astrocytes/pathology , Galectins/metabolism , Humans , Microglia/metabolism , Microglia/pathology , Neurodegenerative Diseases/pathology , Neuroinflammatory Diseases
10.
J Interferon Cytokine Res ; 42(4): 153-160, 2022 04.
Article in English | MEDLINE | ID: mdl-35384725

ABSTRACT

Rapamycin is an immunomodulatory drug that has been evaluated in preclinical and clinical trials as a disease-modifying therapy for multiple sclerosis (MS). In this study, we evaluated the in vitro effect of rapamycin on immune cells pivotally involved in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), which is an animal model to study MS. Splenocytes and central nervous system (CNS)-mononuclear cells obtained from EAE mice were stimulated with a myelin oligodendrocyte glycoprotein peptide, whereas the microglial BV-2 cell line was activated with LPS. The 3 immune cell types were simultaneously treated with rapamycin, incubated, and then used to analyze cytokines, transcription factors, and activation markers. Rapamycin reduced IL-17 production, TBX21, and RORc expression by splenic and CNS cell cultures. IFN-γ and TNF-α production were also decreased in CNS cultures. This treatment also decreased TNF-α, IL-6, MHC II, CD40, and CD86 expression by BV-2 cells. These results indicated that in vivo immunomodulatory activity of rapamycin in MS and EAE was, in many aspects, reproduced by in vitro assays done with cells derived from the spleen and the CNS of EAE mice. This procedure could constitute a screening strategy for choosing drugs with therapeutic potential for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Sirolimus/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
11.
Acta Neuropathol Commun ; 10(1): 31, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35264247

ABSTRACT

Alzheimer's disease (AD) is a major adult-onset neurodegenerative condition with no available treatment. Compelling reports point amyloid-ß (Aß) as the main etiologic agent that triggers AD. Although there is extensive evidence of detrimental crosstalk between Aß and microglia that contributes to neuroinflammation in AD, the exact mechanism leading to neuron death remains unknown. Using postmortem human AD brain tissue, we show that Aß pathology is associated with the necroptosis effector pMLKL. Moreover, we found that the burden of Aß oligomers (Aßo) correlates with the expression of key markers of necroptosis activation. Additionally, inhibition of necroptosis by pharmacological or genetic means, reduce neurodegeneration and memory impairment triggered by Aßo in mice. Since microglial activation is emerging as a central driver for AD pathogenesis, we then tested the contribution of microglia to the mechanism of Aßo-mediated necroptosis activation in neurons. Using an in vitro model, we show that conditioned medium from Aßo-stimulated microglia elicited necroptosis in neurons through activation of TNF-α signaling, triggering extensive neurodegeneration. Notably, necroptosis inhibition provided significant neuronal protection. Together, these findings suggest that Aßo-mediated microglia stimulation in AD contributes to necroptosis activation in neurons and neurodegeneration. As necroptosis is a druggable degenerative mechanism, our findings might have important therapeutic implications to prevent the progression of AD.


Subject(s)
Alzheimer Disease , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Memory Disorders/pathology , Mice , Microglia/pathology , Necroptosis
12.
Sci Rep ; 11(1): 15989, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34362959

ABSTRACT

Different data suggest that microglia may participate in the drug addiction process as these cells respond to neurochemical changes induced by the administration of these substances. In order to study the role of microglia in drug abuse, Swiss mice aged 8-9 weeks were treated with the CSF1R inhibitor PLX3397 (40 mg/kg, p.o.) and submitted to behavioral sensitization or conditioned place preference (CPP) induced by cocaine (15 mg/kg, i.p.). Thereafter, brains were used to evaluate the effects of CSF1R inhibition and cocaine administration on morphological, biochemical and molecular changes. CSF1R inhibition attenuated behavioral sensitization, reduced the number of Iba-1+ cells and increased ramification and lengths of the branches in the remaining microglia. Additionally, both cocaine and PLX3397 increased the cell body to total cell size ratio of Iba-1+ cells, as well as CD68+ and GFAP+ stained areas, suggesting an activated pattern of the glial cells. Besides, CSF1R inhibition increased CX3CL1 levels in the striatum, prefrontal cortex and hippocampus, as well as reduced CX3CR1 expression in the hippocampus. In this region, cocaine also reduced BDNF levels, an effect that was enhanced by CSF1R inhibition. In summary, our results suggest that microglia participate in the behavioral and molecular changes induced by cocaine. This study contributes to the understanding of the role of microglia in cocaine addiction.


Subject(s)
Aminopyridines/pharmacology , Behavior, Animal/drug effects , Cocaine-Related Disorders/prevention & control , Cocaine/toxicity , Microglia/drug effects , Pyrroles/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Cocaine-Related Disorders/etiology , Cocaine-Related Disorders/pathology , Conditioning, Classical , Dopamine Uptake Inhibitors/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Inhibition, Psychological , Male , Mice , Microglia/metabolism , Microglia/pathology
13.
J Immunol Res ; 2021: 7497185, 2021.
Article in English | MEDLINE | ID: mdl-34327244

ABSTRACT

The present study investigated the neuroprotective effect of taurine against the deleterious effects of chronic-recurrent neuroinflammation induced by LPS in the cerebellum of rats. Adult male Wistar rats were treated with taurine for 28 days. Taurine was administered at a dose of 30 or 100 mg/kg, by gavage. On days 7, 14, 21, and 28, the animals received LPS (250 µg/kg) intraperitoneally. The vehicle used was saline. The animals were divided into six groups: vehicle, taurine 30 mg/kg, taurine 100 mg/kg, LPS, LPS plus taurine 30 mg/kg, and LPS plus taurine 100 mg/kg. On day 29, the animals were euthanized, and the cerebellum was removed and prepared for immunofluorescence analysis using antibodies of GFAP, NeuN, CD11b, and cleaved caspase-3. LPS group showed a reduction in the immunoreactivity of GFAP in the arbor vitae and medullary center and of NeuN in the granular layer of the cerebellar cortex. LPS increased the immunoreactivity of CD11b in the arbor vitae and in the medullary center. Taurine protected against these effects induced by LPS in immunoreactivity of GFAP, NeuN, and CD11b, with the 100 mg/kg dose being the most effective. LPS induced an increase in the number of positive cleaved caspase-3 cells in the Purkinje cell layers, granular layer, arbor vitae, and medullary center. Taurine showed its antiapoptotic activity by reducing the cleaved caspase-3 cells in relation to the LPS group. Here, a potential neuroprotective role of taurine can be seen since this amino acid was effective in protecting the cerebellum of rats against cell death and changes in glial and neuronal cells in the face of chronic-recurrent neuroinflammation.


Subject(s)
Cerebellum/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/pharmacology , Taurine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Caspase 3/analysis , Caspase 3/metabolism , Cerebellum/immunology , Cerebellum/pathology , Chronic Disease , Disease Models, Animal , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Male , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neuroinflammatory Diseases/immunology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar , Recurrence , Taurine/therapeutic use
14.
J Biol Chem ; 297(2): 100979, 2021 08.
Article in English | MEDLINE | ID: mdl-34303703

ABSTRACT

Schistosomiasis, a neglected tropical disease caused by trematodes of the Schistosoma genus, affects over 250 million people around the world. This disease has been associated with learning and memory deficits in children, whereas reduced attention levels, impaired work capacity, and cognitive deficits have been observed in adults. Strongly correlated with poverty and lack of basic sanitary conditions, this chronic endemic infection is common in Africa, South America, and parts of Asia and contributes to inhibition of social development and low quality of life in affected areas. Nonetheless, studies on the mechanisms involved in the neurological impairment caused by schistosomiasis are scarce. Here, we used a murine model of infection with Schistosoma mansoni in which parasites do not invade the central nervous system to evaluate the consequences of systemic infection on neurologic function. We observed that systemic infection with S. mansoni led to astrocyte and microglia activation, expression of oxidative stress-induced transcription factor Nrf2, oxidative damage, Tau phosphorylation, and amyloid-ß peptide accumulation in the prefrontal cortex of infected animals. We also found impairment in spatial learning and memory as evaluated by the Morris water maze task. Administration of anthelmintic (praziquantel) and antioxidant (N-acetylcysteine plus deferoxamine) treatments was effective in inhibiting most of these phenotypes, and the combination of both treatments had a synergistic effect to prevent such changes. These data demonstrate new perspectives toward the understanding of the pathology and possible therapeutic approaches to counteract long-term effects of systemic schistosomiasis on brain function.


Subject(s)
Astrocytes/pathology , Microglia/pathology , Neurodegenerative Diseases/pathology , Schistosoma mansoni/isolation & purification , Schistosomiasis mansoni/complications , Acetylcysteine/pharmacology , Animals , Anthelmintics/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Deferoxamine/pharmacology , Disease Models, Animal , Free Radical Scavengers/pharmacology , Male , Mice , Microglia/drug effects , Microglia/metabolism , Morris Water Maze Test/drug effects , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Praziquantel/pharmacology , Schistosoma mansoni/drug effects , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/pathology , Siderophores/pharmacology
15.
Res Vet Sci ; 138: 39-48, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34091228

ABSTRACT

Methotrexate (MTX), an antifolate drug, is widely used in chemotherapeutic protocols for metastatic and primary brain tumors and some autoimmune diseases. Its efficacy for brain tumors is limited by the high incidence of central nervous system (CNS) complications. This investigation aimed to observe the morphological effects, including astroglial and microglial responses, following systemic short-term MTX administration in adult rats. Male Wistar rats received 5 or 10 mg/kg/day of MTX by intraperitoneal route for 4 consecutive days (respectively, MTX5 and MTX10 groups) or the same volume of 0.9% saline solution (control group). On the 5th day, brain samples were collected for hematoxylin-eosin and luxol fast blue staining techniques, as well as for immunohistochemical staining for glial fibrillary acidic protein (GFAP) expression in astrocytes and Iba1 (ionized calcium binding adaptor molecule 1) for microglia in the frontal cortex, hippocampus, hypothalamus and molecular/granular layers of the cerebellum. Morphometric analyses were performed using Image Pro-Plus software. Brain levels of the proinflammatory cytokines TNF-α and IL-1ß were determined by ELISA. No signs of neuronal loss or demyelination were observed in all groups. Increased GFAP and Iba1 expression was found in all areas from the MTX groups, although it was slightly higher in the MTX10 group compared to the MTX5. Both TNF-α and IL-1ß levels were decreased in the MTX5 group compared to controls. In the MTX10 group, TNF-α decreased, although IL-1ß was increased relative to controls. MTX administration induced microglial reaction and astrogliosis in several CNS areas. In the MTX5 group, it apparently occurred in the presence of decreased proinflammatory cytokines.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Astrocytes/drug effects , Calcium-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis/physiopathology , Methotrexate/administration & dosage , Microfilament Proteins/metabolism , Microglia/drug effects , Animals , Astrocytes/metabolism , Astrocytes/pathology , Dose-Response Relationship, Drug , Gliosis/chemically induced , Gliosis/pathology , Male , Microglia/metabolism , Microglia/pathology , Rats , Rats, Wistar
16.
J Neurosci Res ; 99(9): 2287-2304, 2021 09.
Article in English | MEDLINE | ID: mdl-34061383

ABSTRACT

The episodes of cerebral dysfunction, known as encephalopathy, are usually coincident with liver failure. The primary metabolic marker of liver diseases is the increase in blood ammonium, which promotes neuronal damage. In the present project, we used an experimental model of hepatic encephalopathy in male rats by portacaval anastomosis (PCA) surgery. Sham rats had a false operation. After 13 weeks of surgery, the most distinctive finding was vacuolar/spongiform neurodegeneration exclusively in the molecular layer of the cerebellum. This cerebellar damage was further characterized by metabolic, histopathological, and behavioral approaches. The results were as follows: (a) Cellular alterations, namely loss of Purkinje cells, morphological changes, such as swelling of astrocytes and Bergmann glia, and activation of microglia; (b) Cytotoxic edema, shown by an increase in aquaporin-4 and N-acetylaspartate and a reduction in taurine and choline-derivate osmolytes; (c) Metabolic adjustments, noted by the elevation of circulating ammonium, enhanced presence of glutamine synthetase, and increase in glutamine and creatine/phosphocreatine; (d) Inflammasome activation, detected by the elevation of the marker NLRP3 and microglial activation; (e) Locomotor deficits in PCA rats as assessed by the Rotarod and open field tests. These results lead us to suggest that metabolic disturbances associated with PCA can generate the cerebellar damage that is similar to morphophysiological modifications observed in amyloidogenic disorders. In conclusion, we have characterized a distinctive cerebellar multi-disruption accompanied by high levels of ammonium and associated with spongiform neurodegeneration in a model of hepatic hypofunctioning.


Subject(s)
Cerebellum/metabolism , Cerebellum/pathology , Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/pathology , Locomotion/physiology , Portacaval Shunt, Surgical/trends , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cerebellum/surgery , Hepatic Encephalopathy/surgery , Male , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Purkinje Cells/metabolism , Purkinje Cells/pathology , Rats , Rats, Wistar
17.
Int. j. morphol ; 39(3): 920-927, jun. 2021.
Article in Spanish | LILACS | ID: biblio-1385395

ABSTRACT

RESUMEN: El trastorno del espectro autista (TEA) se caracteriza por presentar déficits persistentes en la comunicación y en la interacción social. Además, patrones de comportamiento, intereses o actividades de tipo restrictivo o repetitivo. Su etiología es compleja y heterogenia, y los mecanismos neurobiológicos que dan lugar al fenotipo clínico aún no se conocen por completo. Las investigaciones apuntan a factores genéticos y ambientales que afectan el cerebro en desarrollo. Estos avances coinciden con un aumento en la comprensión de las funciones fisiológicas y el potencial patológico de la neuroglia en el sistema nervioso central (SNC) que llevó a la noción de la contribución fundamental de estas células en el TEA. Así, el objetivo de este artículo fue revisar brevemente los factores de riesgo clave asociados al TEA y luego, explorar la contribución de la neuroglia en este trastorno. Se destaca el rol de los astrocitos, los microglocitos y los oligodendrocitos en el control homeostático del SNC, en la regulación inmunitaria del cerebro y en la mielinización axonal, así como el mal funcionamiento y las alteraciones morfológicas de estas células en los cerebros autistas.


SUMMARY: Autism spectrum disorder (ASD) is characterized by persistent deficits in communication and social interaction, as well as restrictive or repetitive activities or interests. Its etiology is complex and heterogeneous, and the neurobiological mechanisms that give rise to the clinical phenotype are not yet fully understood. Research points to genetic and environmental factors that affect the developing brain. These advances are consistent with an enhanced understanding of the physiological functions and pathological potential of neuroglia in the central nervous system (CNS) which supports the conclusion of the contribution of these cells in ASD. Therefore, the objective of this article was to briefly review the key risk factors associated with ASD and then explore the contribution of glia in this disorder. The role of astrocytes, microgliocytes and oligodendrocytes in the homeostatic control of the CNS in the immune regulation of the brain and in axonal myelination, as well as malfunction and morphological alterations of these cells in autistic brains are emphasized.


Subject(s)
Humans , Neuroglia/pathology , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/pathology , Oligodendroglia/pathology , Astrocytes/pathology , Microglia/pathology , Autism Spectrum Disorder/etiology , Homeostasis
18.
J Biosci ; 462021.
Article in English | MEDLINE | ID: mdl-34047285

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative condition provoking the loss of cognitive and memory performances. Despite huge efforts to develop effective AD therapies, there is still no cure for this neurological condition. Here, we review the main biological properties of Phycocyanobilin (PCB), accounting for its potential uses against AD. PCB, given individually or released in vivo from C-Phycocyanin (C-PC), acts as a bioactive-molecule-mediating antioxidant, is anti-inflammatory and has immunomodulatory activities. PCB/C-PC are able to scavenge reactive oxygen and nitrogen species, to counteract lipid peroxidation and to inhibit enzymes such as NADPH oxidase and COX-2. In animal models of multiple sclerosis and ischemic stroke, these compounds induce remyelination as demonstrated by electron microscopy and the expression of genes such as Mal up-regulation of and Lingo-1 down-regulation. These treatments also reduce pro-inflammatory cytokines levels and induce immune suppressive genes. PCB/C-PC protects isolated rat brain mitochondria and inactivate microglia, astrocytes and neuronal apoptosis mediators. Such processes are all involved in the pathogenic cascade of AD, and thus PCB may effectively mitigate the injury in this condition. Furthermore, PCB can be administered safely by oral or parenteral routes and therefore, could be commercially offered as a nutraceutical supplement or as a pharmaceutical drug.


Subject(s)
Alzheimer Disease/drug therapy , Dietary Supplements/analysis , Phycobilins/pharmacology , Phycocyanin/pharmacology , Remyelination/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Immunologic Factors/pharmacology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oxidative Stress/drug effects , Rats , Reactive Nitrogen Species , Reactive Oxygen Species
19.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33915754

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease, characterized histopathologically by intra-neuronal tau-related lesions and by the accumulation of amyloid ß-peptide (Aß) in the brain parenchyma and around cerebral blood vessels. According to the vascular hypothesis of AD, an alteration in the neurovascular unit (NVU) could lead to Aß vascular accumulation and promote neuronal dysfunction, accelerating neurodegeneration and dementia. To date, the effects of insoluble vascular Aß deposits on the NVU and the blood-brain barrier (BBB) are unknown. In this study, we analyze different Aß species and their association with the cells that make up the NVU. We evaluated post-mortem AD brain tissue. Multiple immunofluorescence assays were performed against different species of Aß and the main elements that constitute the NVU. Our results showed that there are insoluble vascular deposits of both full-length and truncated Aß species. Besides, insoluble aggregates are associated with a decrease in the phenotype of the cellular components that constitute the NVU and with BBB disruption. This approach could help identify new therapeutic targets against key molecules and receptors in the NVU that can prevent the accumulation of vascular fibrillar Aß in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Astrocytes/pathology , Blood Vessels/pathology , Brain/pathology , Microglia/pathology , Actins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Blood Vessels/metabolism , Brain/blood supply , Brain/metabolism , Case-Control Studies , Caspases/metabolism , Humans , Tight Junctions/pathology
20.
Int J Mol Sci ; 22(5)2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33668314

ABSTRACT

Under high-fat feeding, the hypothalamus atypically undergoes pro-inflammatory signaling activation. Recent data from transcriptomic analysis of microglia from rodents and humans has allowed the identification of several microglial subpopulations throughout the brain. Numerous studies have clarified the roles of these cells in hypothalamic inflammation, but how each microglial subset plays its functions upon inflammatory stimuli remains unexplored. Fortunately, these data unveiling microglial heterogeneity have triggered the development of novel experimental models for studying the roles and characteristics of each microglial subtype. In this review, we explore microglial heterogeneity in the hypothalamus and their crosstalk with astrocytes under high fat diet-induced inflammation. We present novel currently available ex vivo and in vivo experimental models that can be useful when designing a new research project in this field of study. Last, we examine the transcriptomic data already published to identify how the hypothalamic microglial signature changes upon short-term and prolonged high-fat feeding.


Subject(s)
Astrocytes/pathology , Diet, High-Fat/adverse effects , Hypothalamus/pathology , Inflammation/pathology , Microglia/pathology , Transcriptome , Animals , Astrocytes/metabolism , Humans , Hypothalamus/metabolism , Inflammation/etiology , Inflammation/metabolism , Microglia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL