Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 107: 153-164, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36202169

RESUMO

Sleep is a natural physiological state, tightly regulated through several neuroanatomical and neurochemical systems, which is essential to maintain physical and mental health. Recent studies revealed that the functions of microglia, the resident immune cells of the brain, differ along the sleep-wake cycle. Inflammatory cytokines, such as interleukin-1ß and tumor necrosis factor-α, mainly produced by microglia in the brain, are also well-known to promote sleep. However, the contributing role of microglia on sleep regulation remains largely elusive, even more so in females. Given the higher prevalence of various sleep disorders in women, we aimed to determine the role of microglia in regulating the sleep-wake cycle specifically in female mice. Microglia were depleted in adult female mice with inhibitors of the colony-stimulating factor 1 receptor (CSF1R) (PLX3397 or PLX5622), which is required for microglial population maintenance. This led to a 65-73% reduction of the microglial population, as confirmed by immunofluorescence staining against IBA1 (marker of microglia/macrophages) and TMEM119 (microglia-specific marker) in the reticular nucleus of the thalamus and primary motor cortex. The spontaneous sleep-wake cycle was evaluated at steady-state, during microglial homeostasis disruption and after complete microglial repopulation, upon cessation of treatment with the inhibitors of CSF1R, using electroencephalography (EEG) and electromyography (EMG). We found that microglia-depleted female mice spent more time in non-rapid eye movement (NREM) sleep and had an increased number of NREM sleep episodes, which was partially restored after microglial total repopulation. To determine whether microglia could regulate sleep locally by modulating synaptic transmission, we used patch clamp to record spontaneous activity of pyramidal neurons in the primary motor cortex, which showed an increase of excitatory synaptic transmission during the dark phase. These changes in neuronal activity were modulated by microglial depletion in a phase-dependent manner. Altogether, our results indicate that microglia are involved in the sleep regulation of female mice, further strengthening their potential implication in the development and/or progression of sleep disorders. Furthermore, our findings indicate that microglial repopulation can contribute to normalizing sleep alterations caused by their partial depletion.


Assuntos
Movimentos Oculares , Transtornos do Sono-Vigília , Feminino , Animais , Camundongos , Duração do Sono , Fator de Necrose Tumoral alfa
2.
Micron ; 161: 103334, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35970079

RESUMO

Microglia, the immune resident cells of the central nervous system (CNS), are now recognized as performing crucial roles for maintaining homeostasis and determining the outcomes of various pathological challenges across life. While brightfield microscopy is a powerful and established tool to study microglia-mediated mechanisms underlying neurological diseases, microglial density and distribution are some of the most frequently investigated parameters. Their quantitative assessment provides relevant clues regarding dynamic densitometric changes in the microglial population across various CNS regions. Investigators often rely on a manual identification and analysis of these cells within key regions of interest, which can be time-consuming and introduce an experimenter bias. Automation of this process, which has been gaining popularity in recent years, represents a potential solution to minimize both experimenter's bias and time investment, thus increasing the efficacy of the experiment and uniformity of the collected data. We aimed to compare manual versus automatic analysis methods to determine whether an automatic analysis is efficient and accurate enough to replace a manual analysis in both homeostatic and pathological contexts (i.e., adult healthy and lipopolysaccharide-challenged adolescent male mice, respectively). To do so, we used a script that runs on the ImageJ software to perform microglial density analysis by automatic detection of microglial cells from brightfield microscopy images. The main core of the macro script consists in an automatic cell selection step using a threshold followed by a spatial analysis for each selected cell. The resulting data were then compared with the values obtained using a well-established manual method. Overall, the evaluation of the established automatic densitometry method with manual density and distribution analysis revealed similar results for the density and nearest neighbor distance in healthy adult mice, as well as density and distribution in lipopolysaccharide-challenged adolescent mice. Applying machine learning to the automatic process could further improve the accuracy and robustness of the method.


Assuntos
Lipopolissacarídeos , Microglia , Animais , Camundongos , Masculino , Microglia/patologia , Hipocampo , Software , Automação
3.
J Neuroinflammation ; 19(1): 81, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-35387656

RESUMO

BACKGROUND: Microglia participate in the immune response upon central nervous system (CNS) infections. However, the role of these cells during herpes simplex encephalitis (HSE) has not been fully characterized. We sought to identify different microglia/microglia-like cells and describe the potential mechanisms and signaling pathways involved during HSE. METHODS: The transcriptional response of CD11b+ immune cells, including microglia/microglia-like cells, was investigated using single-cell RNA sequencing (scRNA-seq) on cells isolated from the ventral posterolateral nucleus (VPL)-enriched thalamic regions of C57BL/6 N mice intranasally infected with herpes simplex virus-1 (HSV-1) (6 × 105 PFUs/20 µl). We further performed scanning electronic microscopy (SEM) analysis in VPL regions on day 6 post-infection (p.i.) to provide insight into microglial functions. RESULTS: We describe a novel microglia-like transcriptional response associated with a rare cell population (7% of all analyzed cells), named "in transition" microglia/microglia-like cells in HSE. This new microglia-like transcriptional signature, found in the highly infected thalamic regions, was enriched in specific genes (Retnlg, Cxcr2, Il1f9) usually associated with neutrophils. Pathway analysis of this cell-type transcriptome showed increased NLRP3-inflammasome-mediated interleukin IL-1ß production, promoting a pro-inflammatory response. These cells' increased expression of viral transcripts suggests that the distinct "in transition" transcriptome corresponds to the intrinsic antiviral immune signaling of HSV-1-infected microglia/microglia-like cells in the thalamus. In accordance with this phenotype, we observed several TMEM119+/IBA-I+ microglia/microglia-like cells immunostained for HSV-1 in highly infected regions. CONCLUSIONS: A new microglia/microglia-like state may potentially shed light on how microglia could react to HSV-1 infection. Our observations suggest that infected microglia/microglia-like cells contribute to an exacerbated CNS inflammation. Further characterization of this transitory state of the microglia/microglia-like cell transcriptome may allow the development of novel immunomodulatory approaches to improve HSE outcomes by regulating the microglial immune response.


Assuntos
Encefalite por Herpes Simples , Herpesvirus Humano 1 , Animais , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Transcriptoma , Núcleos Ventrais do Tálamo
4.
Proc Natl Acad Sci U S A ; 119(12): e2114545119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35286203

RESUMO

Exposure to maternal immune activation (MIA) in utero is a risk factor for neurodevelopmental and psychiatric disorders. MIA-induced deficits in adolescent and adult offspring have been well characterized; however, less is known about the effects of MIA exposure on embryo development. To address this gap, we performed high-resolution ex vivo MRI to investigate the effects of early (gestational day [GD]9) and late (GD17) MIA exposure on embryo (GD18) brain structure. We identify striking neuroanatomical changes in the embryo brain, particularly in the late-exposed offspring. We further examined the putative neuroanatomical underpinnings of MIA timing in the hippocampus using electron microscopy and identified differential effects due to MIA timing. An increase in apoptotic cell density was observed in the GD9-exposed offspring, while an increase in the density of neurons and glia with ultrastructural features reflective of increased neuroinflammation and oxidative stress was observed in GD17-exposed offspring, particularly in females. Overall, our findings integrate imaging techniques across different scales to identify differential impact of MIA timing on the earliest stages of neurodevelopment.


Assuntos
Transtorno do Espectro Autista , Sistema Imunitário , Efeitos Tardios da Exposição Pré-Natal , Esquizofrenia , Adolescente , Animais , Encéfalo , Modelos Animais de Doenças , Feminino , Humanos , Sistema Imunitário/fisiologia , Inflamação , Imageamento por Ressonância Magnética , Camundongos , Gravidez
5.
Commun Biol ; 5(1): 26, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017640

RESUMO

Various environmental exposures during pregnancy, like maternal diet, can compromise, at critical periods of development, the neurovascular maturation of the offspring. Foetal exposure to maternal high-fat diet (mHFD), common to Western societies, has been shown to disturb neurovascular development in neonates and long-term permeability of the neurovasculature. Nevertheless, the effects of mHFD on the offspring's cerebrovascular health remains largely elusive. Here, we sought to address this knowledge gap by using a translational mouse model of mHFD exposure. Three-dimensional and ultrastructure analysis of the neurovascular unit (vasculature and parenchymal cells) in mHFD-exposed offspring revealed major alterations of the neurovascular organization and metabolism. These alterations were accompanied by changes in the expression of genes involved in metabolism and immunity, indicating that neurovascular changes may result from abnormal brain metabolism and immune regulation. In addition, mHFD-exposed offspring showed persisting behavioural alterations reminiscent of neurodevelopmental disorders, specifically an increase in stereotyped and repetitive behaviours into adulthood.


Assuntos
Comportamento Animal/fisiologia , Córtex Cerebral , Dieta Hiperlipídica/efeitos adversos , Exposição Materna , Microglia/patologia , Animais , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/citologia , Córtex Cerebral/patologia , Feminino , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal
6.
Glia ; 70(1): 50-70, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34519378

RESUMO

Westernization of dietary habits has led to a progressive reduction in dietary intake of n-3 polyunsaturated fatty acids (n-3 PUFAs). Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental disorders, conditions in which myelination processes are abnormal, leading to defects in brain functional connectivity. Only little is known about the role of n-3 PUFAs in oligodendrocyte physiology and white matter development. Here, we show that lifelong n-3 PUFA deficiency disrupts oligodendrocytes maturation and myelination processes during the postnatal period in mice. This has long-term deleterious consequences on white matter organization and hippocampus-prefrontal functional connectivity in adults, associated with cognitive and emotional disorders. Promoting developmental myelination with clemastine, a first-generation histamine antagonist and enhancer of oligodendrocyte precursor cell differentiation, rescues memory deficits in n-3 PUFA deficient animals. Our findings identify a novel mechanism through which n-3 PUFA deficiency alters brain functions by disrupting oligodendrocyte maturation and brain myelination during the neurodevelopmental period.


Assuntos
Ácidos Graxos Ômega-3 , Animais , Encéfalo , Camundongos , Bainha de Mielina , Neurogênese , Oligodendroglia
7.
Brain Behav Immun Health ; 15: 100281, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34589781

RESUMO

Prenatal exposure to maternal high-fat diet (mHFD) acts as a risk factor for various neurodevelopmental alterations in the progeny. Recent studies in mice revealed that mHFD results in both neuroinflammation and hypomyelination in the exposed offspring. Microglia, the brain-resident macrophages, play crucial roles during brain development, notably by modulating oligodendrocyte populations and performing phagocytosis of myelin sheaths. Previously, we reported that mHFD modifies microglial phenotype (i.e., morphology, interactions with their microenvironment, transcripts) in the hippocampus of male and female offspring. In the current study, we further explored whether mHFD may induce myelination changes among the hippocampal-corpus callosum-prefrontal cortex pathway, and result in behavioral outcomes in adolescent offspring of the two sexes. To this end, female mice were fed with control chow or HFD for 4 weeks before mating, during gestation, and until weaning of their litter. Histological and ultrastructural analyses revealed an increased density of myelin associated with a reduced area of cytosolic myelin channels in the corpus callosum of mHFD-exposed male compared to female offspring. Transcripts of myelination-associated genes including Igf1 -a growth factor released by microglia- were also lower, specifically in the hippocampus (without changes in the prefrontal cortex) of adolescent male mouse offspring. These changes in myelin were not related to an altered density, distribution, or maturation of oligodendrocytes, instead we found that microglia within the corpus callosum of mHFD-exposed offspring showed reduced numbers of mature lysosomes and increased synaptic contacts, suggesting microglial implication in the modified myelination. At the behavioral level, both male and female mHFD-exposed adolescent offspring presented loss of social memory and sensorimotor gating deficits. These results together highlight the importance of studying oligodendrocyte-microglia crosstalk and its involvement in the long-term brain alterations that result from prenatal mHFD in offspring across sexes.

8.
J Neuroinflammation ; 18(1): 178, 2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34399779

RESUMO

BACKGROUND: Zika virus (ZIKV) has been associated with several neurological complications in adult patients. METHODS: We used a mouse model deficient in TRIF and IPS-1 adaptor proteins, which are involved in type I interferon production, to study the role of microglia during brain infection by ZIKV. Young adult mice were infected intravenously with the contemporary ZIKV strain PRVABC59 (1 × 105 PFUs/100 µL). RESULTS: Infected mice did not present overt clinical signs of the disease nor body weight loss compared with noninfected animals. However, mice exhibited a viremia and a brain viral load that were maximal (1.3 × 105 genome copies/mL and 9.8 × 107 genome copies/g of brain) on days 3 and 7 post-infection (p.i.), respectively. Immunohistochemistry analysis showed that ZIKV antigens were distributed in several regions of the brain, especially the dorsal hippocampus. The number of Iba1+/TMEM119+ microglia remained similar in infected versus noninfected mice, but their cell body and arborization areas significantly increased in the stratum radiatum and stratum lacunosum-moleculare layers of the dorsal hippocampus cornu ammoni (CA)1, indicating a reactive state. Ultrastructural analyses also revealed that microglia displayed increased phagocytic activities and extracellular digestion of degraded elements during infection. Mice pharmacologically depleted in microglia with PLX5622 presented a higher brain viral load compared to untreated group (2.8 × 1010 versus 8.5 × 108 genome copies/g of brain on day 10 p.i.) as well as an increased number of ZIKV antigens labeled with immunogold in the cytoplasm and endoplasmic reticulum of neurons and astrocytes indicating an enhanced viral replication. Furthermore, endosomes of astrocytes contained nanogold particles together with digested materials, suggesting a compensatory phagocytic activity upon microglial depletion. CONCLUSIONS: These results indicate that microglia are involved in the control of ZIKV replication and/or its elimination in the brain. After depletion of microglia, the removal of ZIKV-infected cells by phagocytosis could be partly compensated by astrocytes.


Assuntos
Encéfalo/virologia , Microglia/metabolismo , Neurônios/metabolismo , Fagocitose/fisiologia , Infecção por Zika virus/metabolismo , Animais , Encéfalo/metabolismo , Camundongos , Microglia/virologia , Neurônios/virologia
9.
Brain Behav Immun ; 97: 440-454, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34343619

RESUMO

Inflammation during pregnancy can disturb brain development and lead to disorders in the progeny, including autism spectrum disorder and schizophrenia. However, the mechanism by which a prenatal, short-lived increase of cytokines results in adverse neurodevelopmental outcomes remains largely unknown. Microglia-the brain's resident immune-cells-stand as fundamental cellular mediators, being highly sensitive and responsive to immune signals, which also play key roles during normal development. The fractalkine signaling axis is a neuron-microglia communication mechanism used to regulate neurogenesis and network formation. Previously, we showed hippocampal reduction of fractalkine receptor (Cx3cr1) mRNA at postnatal day (P) 15 in male offspring exposed to maternal immune activation induced with lipopolysaccharide (LPS) during late gestation, which was concomitant to an increased dendritic spine density in the dentate gyrus, a neurogenic niche. The current study sought to evaluate the origin and impact of this reduced hippocampal Cx3cr1 mRNA expression on microglia and cognition. We found that microglial total cell number and density are not affected in the dorsal hippocampus and dentate gyrus, respectively, but that the microglial CX3CR1 protein is decreased in the hippocampus of LPS-male offspring at P15. Further characterization of microglial morphology in the dentate gyrus identified a more ameboid phenotype in LPS-exposed offspring, predominantly in males, at P15. We thus explored maternal plasma and fetal brain cytokines to understand the mechanism behind microglial priming, showing a robust immune activation in the mother at 2 and 4 hrs after LPS administration, while only IL-10 tended towards upregulation at 2 hrs after LPS in fetal brains. To evaluate the functional long-term consequences, we assessed learning and cognitive flexibility behavior during late adolescence, finding that LPS affects only the latter with a male predominance on perseveration. A CX3CR1 gene variant in humans that results in disrupted fractalkine signaling has been recently associated with an increased risk for neurodevelopmental disorders. We show that an acute immune insult during late gestation can alter fractalkine signaling by reducing the microglial CX3CR1 protein expression, highlighting neuron-microglial fractalkine signaling as a relevant target underlying the outcomes of environmental risk factors on neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista , Receptor 1 de Quimiocina CX3C/genética , Microglia , Adolescente , Receptor 1 de Quimiocina CX3C/metabolismo , Cognição , Giro Denteado/metabolismo , Feminino , Hipocampo/metabolismo , Humanos , Lipopolissacarídeos , Masculino , Microglia/metabolismo , Fenótipo , Gravidez
10.
Neurochem Int ; 145: 104987, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33587954

RESUMO

Microglia, the resident macrophage cells of the central nervous system (CNS), are involved in a myriad of processes required to maintain CNS homeostasis. These cells are dynamic and can adapt their phenotype and functions to the physiological needs of the organism. Microglia rapidly respond to changes occurring in their microenvironment, such as the ones taking place during stress. While stress can be beneficial for the organism to adapt to a situation, it can become highly detrimental when it turns chronic. Microglial response to prolonged stress may lead to an alteration of their beneficial physiological functions, becoming either maladaptive or pro-inflammatory. In this review, we aim to summarize the effects of chronic stress exerted on microglia through the neuroendocrine system and inflammation at adulthood. We also discuss how these effects of chronic stress could contribute to microglial involvement in neuropsychiatric and sleep disorders, as well as neurodegenerative diseases.


Assuntos
Mediadores da Inflamação/metabolismo , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Sistemas Neurossecretores/metabolismo , Estresse Psicológico/metabolismo , Animais , Doença Crônica , Corticosterona/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Microglia/patologia , Doenças Neurodegenerativas/patologia , Sistemas Neurossecretores/patologia , Norepinefrina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Psicológico/patologia
11.
Front Cell Neurosci ; 14: 592214, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33304243

RESUMO

SARS-CoV-2, which causes the Coronavirus Disease 2019 (COVID-19) pandemic, has a brain neurotropism through binding to the receptor angiotensin-converting enzyme 2 expressed by neurones and glial cells, including astrocytes and microglia. Systemic infection which accompanies severe cases of COVID-19 also triggers substantial increase in circulating levels of chemokines and interleukins that compromise the blood-brain barrier, enter the brain parenchyma and affect its defensive systems, astrocytes and microglia. Brain areas devoid of a blood-brain barrier such as the circumventricular organs are particularly vulnerable to circulating inflammatory mediators. The performance of astrocytes and microglia, as well as of immune cells required for brain health, is considered critical in defining the neurological damage and neurological outcome of COVID-19. In this review, we discuss the neurotropism of SARS-CoV-2, the implication of neuroinflammation, adaptive and innate immunity, autoimmunity, as well as astrocytic and microglial immune and homeostatic functions in the neurological and psychiatric aspects of COVID-19. The consequences of SARS-CoV-2 infection during ageing, in the presence of systemic comorbidities, and for the exposed pregnant mother and foetus are also covered.

12.
J Neuroinflammation ; 17(1): 264, 2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32891154

RESUMO

BACKGROUND: Maternal nutrition is critical for proper fetal development. While increased nutrient intake is essential during pregnancy, an excessive consumption of certain nutrients, like fat, can lead to long-lasting detrimental consequences on the offspring. Animal work investigating the consequences of maternal high-fat diet (mHFD) revealed in the offspring a maternal immune activation (MIA) phenotype associated with increased inflammatory signals. This inflammation was proposed as one of the mechanisms causing neuronal circuit dysfunction, notably in the hippocampus, by altering the brain-resident macrophages-microglia. However, the understanding of mechanisms linking inflammation and microglial activities to pathological brain development remains limited. We hypothesized that mHFD-induced inflammation could prime microglia by altering their specific gene expression signature, population density, and/or functions. METHODS: We used an integrative approach combining molecular (i.e., multiplex-ELISA, rt-qPCR) and cellular (i.e., histochemistry, electron microscopy) techniques to investigate the effects of mHFD (saturated and unsaturated fats) vs control diet on inflammatory priming, as well as microglial transcriptomic signature, density, distribution, morphology, and ultrastructure in mice. These analyses were performed on the mothers and/or their adolescent offspring at postnatal day 30. RESULTS: Our study revealed that mHFD results in MIA defined by increased circulating levels of interleukin (IL)-6 in the mothers. This phenotype was associated with an exacerbated inflammatory response to peripheral lipopolysaccharide in mHFD-exposed offspring of both sexes. Microglial morphology was also altered, and there were increased microglial interactions with astrocytes in the hippocampus CA1 of mHFD-exposed male offspring, as well as decreased microglia-associated extracellular space pockets in the same region of mHFD-exposed offspring of the two sexes. A decreased mRNA expression of the inflammatory-regulating cytokine Tgfb1 and microglial receptors Tmem119, Trem2, and Cx3cr1 was additionally measured in the hippocampus of mHFD-exposed offspring, especially in males. CONCLUSIONS: Here, we described how dietary habits during pregnancy and nurturing, particularly the consumption of an enriched fat diet, can influence peripheral immune priming in the offspring. We also found that microglia are affected in terms of gene expression signature, morphology, and interactions with the hippocampal parenchyma, in a partially sexually dimorphic manner, which may contribute to the adverse neurodevelopmental outcomes on the offspring.


Assuntos
Dieta Hiperlipídica , Hipocampo/patologia , Inflamação/patologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Microglia/patologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Adolescente , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Forma Celular/fisiologia , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-6/sangue , Lipopolissacarídeos/farmacologia , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Gravidez , Receptores Imunológicos/metabolismo , Fatores Sexuais , Fator de Crescimento Transformador beta1/metabolismo
13.
Brain Behav Immun ; 90: 81-96, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32755645

RESUMO

Parkinson's disease (PD) is the most common neurodegenerative motor disorder. The mechanisms underlying the onset and progression of Levodopa (L-Dopa)-induced dyskinesia (LID) during PD treatment remain elusive. Emerging evidence implicates functional modification of microglia in the development of LID. Thus, understanding the link between microglia and the development of LID may provide the knowledge required to preserve or promote beneficial microglial functions, even during a prolonged L-Dopa treatment. To provide novel insights into microglial functional alterations in PD pathophysiology, we characterized their density, morphology, ultrastructure, and degradation activity in the sensorimotor functional territory of the putamen, using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) cynomolgus monkeys. A subset of MPTP monkeys was treated orally with L-Dopa and developed LID similar to PD patients. Using a combination of light, confocal and transmission electron microscopy, our quantitative analyses revealed alterations of microglial density, morphology and phagolysosomal activity following MPTP intoxication that were partially normalized with L-Dopa treatment. In particular, microglial density, cell body and arborization areas were increased in the MPTP monkeys, whereas L-Dopa-treated MPTP animals presented a microglial phenotype similar to the control animals. At the ultrastructural level, microglia did not differ between groups in their markers of cellular stress or aging. Nevertheless, microglia from the MPTP monkeys displayed reduced numbers of endosomes, compared with control animals, that remained lower after L-Dopa treatment. Microglia from MPTP monkeys treated with L-Dopa also had increased numbers of primary lysosomes compared with non-treated MPTP animals, while secondary and tertiary lysosomes remained unchanged. Moreover, a decrease microglial immunoreactivity for CD68, considered a marker of phagocytosis and lysosomal activity, was measured in the MPTP monkeys treated with L-Dopa, compared with non-treated MPTP animals. Taken together, these findings revealed significant changes in microglia during PD pathophysiology that were partially rescued by L-Dopa treatment. Albeit, this L-Dopa treatment conferred phagolysosomal insufficiency on microglia in the dyskinetic Parkinsonian monkeys.


Assuntos
Discinesia Induzida por Medicamentos , Doença de Parkinson , Animais , Discinesia Induzida por Medicamentos/tratamento farmacológico , Humanos , Levodopa , Macaca fascicularis , Microglia , Doença de Parkinson/tratamento farmacológico
14.
Front Cell Neurosci ; 14: 612705, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33536875

RESUMO

Providing the appropriate quantity and quality of food needed for both the mother's well-being and the healthy development of the offspring is crucial during pregnancy. However, the macro- and micronutrient intake also impacts the body's regulatory supersystems of the mother, such as the immune, endocrine, and nervous systems, which ultimately influence the overall development of the offspring. Of particular importance is the association between unhealthy maternal diet and neurodevelopmental disorders in the offspring. Epidemiological studies have linked neurodevelopmental disorders like autism spectrum disorders, attention-deficit-hyperactivity disorder, and schizophrenia, to maternal immune activation (MIA) during gestation. While the deleterious consequences of diet-induced MIA on offspring neurodevelopment are increasingly revealed, neuroinflammation is emerging as a key underlying mechanism. In this review, we compile the evidence available on how the mother and offspring are both impacted by maternal dietary imbalance. We specifically explore the various inflammatory and anti-inflammatory effects of dietary components and discuss how changes in inflammatory status can prime the offspring brain development toward neurodevelopmental disorders. Lastly, we discuss research evidence on the mechanisms that sustain the relationship between maternal dietary imbalance and offspring brain development, involving altered neuroinflammatory status in the offspring, as well as genetic to cellular programming notably of microglia, and the evidence that the gut microbiome may act as a key mediator.

15.
J Vis Exp ; (152)2019 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-31710033

RESUMO

This is a protocol for the dual visualization of microglia and infiltrating macrophages in mouse brain tissue. TMEM119 (which labels microglia selectively), when combined with IBA1 (which provides an exceptional visualization of their morphology), allows investigation of changes in density, distribution, and morphology. Quantifying these parameters is important in providing insights into the roles exerted by microglia, the resident macrophages of the brain. Under normal physiological conditions, microglia are regularly distributed in a mosaic-like pattern and present a small soma with ramified processes. Nevertheless, as a response to environmental factors (i.e., trauma, infection, disease, or injury), microglial density, distribution, and morphology are altered in various manners, depending on the insult. Additionally, the described double-staining method allows visualization of infiltrating macrophages in the brain based on their expression of IBA1 and without colocalization with TMEM119. This approach thus allows discrimination between microglia and infiltrating macrophages, which is required to provide functional insights into their distinct involvement in brain homeostasis across various contexts of health and disease. This protocol integrates the latest findings in neuroimmunology that pertain to the identification of selective markers. It also serves as a useful tool for both experienced neuroimmunologists and researchers seeking to integrate neuroimmunology into projects.


Assuntos
Encéfalo/fisiologia , Imunofluorescência/métodos , Macrófagos/fisiologia , Microglia/fisiologia , Células Mieloides/fisiologia , Coloração e Rotulagem/métodos , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Homeostase , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Microglia/citologia , Microglia/metabolismo , Células Mieloides/citologia , Células Mieloides/metabolismo
16.
Methods Mol Biol ; 2034: 97-110, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31392680

RESUMO

Dark microglia, a recently described phenotype, are found in high numbers in nonhomeostatic conditions (e.g., Alzheimer's disease pathology, aging, chronic stress). As a specific protein marker has not yet been defined, they cannot be studied using conventional cellular biology techniques. They are recognized by their unique ultrastructural features visible under electron microscopy. This nanoscale resolution imaging technique allows the identification of cells based on their ultrastructure or immunoreactivity to certain proteins. In this protocol, we describe the steps necessary for the preparation of high-quality brain tissues for transmission electron microscopy, the imaging, the identification of dark microglia, and the ultrastructural analysis of various parameters that can be studied in these cells.


Assuntos
Doença de Alzheimer , Encéfalo , Microglia , Microscopia Eletrônica de Transmissão , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/ultraestrutura , Microglia/metabolismo , Microglia/ultraestrutura
17.
Semin Cell Dev Biol ; 94: 152-163, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31201858

RESUMO

In addition to their traditional role as immune sentinels, recent discoveries over the last decade have shown that microglial functions now include regulation of neuronal/glial cell migration, differentiation and maturation, as well as neuronal network formation. It was thus proposed that disruption of these microglial roles, during critical periods of brain development, could lead to the pathological onset of several neurodevelopmental disorders, including autism spectrum disorder, attention deficit hyperactivity disorder, epilepsy, schizophrenia, and major depressive disorder. The prevalence of these disorders exhibits a clear distinction along sex lines with very little known about the mechanisms underlying this difference. One of the fundamental discoveries that arose from recent research into the physiological roles of microglia in neurodevelopment is their sexual dimorphism, raising the intriguing possibility that sex differences in microglial colonization, maturation and/or function in the developing brain could underlie the emergence of various neurodevelopmental disorders. This review discusses the physiological roles of microglia across neurodevelopment, these roles in the two sexes, and the recent evidence that microglial sexually dimorphic nature may contribute, at least partially, to neurodevelopmental disorders.


Assuntos
Encéfalo/citologia , Encéfalo/fisiologia , Microglia/patologia , Microglia/fisiologia , Transtornos do Neurodesenvolvimento/patologia , Caracteres Sexuais , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Microglia/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo
18.
Front Cell Neurosci ; 12: 282, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30214398

RESUMO

Microglia, often described as the brain-resident macrophages, play crucial roles in central nervous system development, maintenance, plasticity, and adaptation to the environment. Both aging and chronic stress promote microglial morphological and functional changes, which can lead to the development of brain pathologies including Parkinson's disease (PD). Indeed, aging, and chronic stress represent main environmental risk factors for PD. In these conditions, microglia are known to undergo different morphological and functional changes. Inflammation is an important component of PD and disequilibrium between pro- and anti-inflammatory microglial functions might constitute a crucial component of PD onset and progression. Cumulated data also suggest that, during PD, microglia might lose beneficial functions and gain detrimental ones, in addition to mediating inflammation. In this mini-review, we aim to summarize the literature discussing the functional and morphological changes that microglia undergo in PD pathophysiology and upon exposure to its two main environmental risk factors, aging, and chronic stress.

19.
Oncotarget ; 7(11): 11864-80, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26918610

RESUMO

Cerebral vasculature plays a key role in controlling brain homeostasis. Cerebral vasculature dysfunction, associated to irregularities in cerebral blood perfusion, has been proposed to directly contribute to Alzheimer's disease (AD) pathogenesis. More precisely, chronic cerebral hypoperfusion, which impairs brain homeostasis, was demonstrated to take place even before cognitive decline. However, the mechanisms underlying the implication of chronic cerebral hypoperfusion in AD pathogenesis remain elusive. Therefore, this study aims at investigating the role of severe chronic cerebral hypoperfusion (SCCH) in AD pathogenesis. For this purpose, SCCH was induced in young APPswe/PS1 in order to evaluate the progression of AD-like pathology in these mice. We observed that SCCH accelerated the cognitive decline of young APPswe/PS1 mice, which was associated with an increased amyloid plaque number in brain parenchyma. In addition, SCCH reduced the activity of extracellular signal-regulated kinases 1/2 (ERK1/2), which has been shown to play an important role in the adaptive responses of neurons. Importantly, SCCH impaired the function of microglial cells, which are implicated in amyloid-ß (Aß) elimination. In vitro approaches underlined the ability of a low-glucose microenvironment to decrease the general activity and phagocytic capacity of microglia. By using a new model of SCCH, our study unravels new insights into the implication of severe chronic cerebral hypoperfusion in AD pathogenesis, mainly by altering microglial cell activity and consequently Aß clearance.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Transtornos Cerebrovasculares/complicações , Transtornos Cognitivos/etiologia , Transtornos da Memória/etiologia , Microglia/patologia , Presenilina-1/fisiologia , Índice de Gravidade de Doença , Animais , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Circulação Cerebrovascular , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Doença Crônica , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Humanos , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Microglia/metabolismo
20.
Neuropsychopharmacology ; 41(5): 1297-307, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26349911

RESUMO

Alzheimer's disease (AD) is the leading cause of dementia among elderly population. AD is characterized by the accumulation of beta-amyloid (Aß) peptides, which aggregate over time to form amyloid plaques in the brain. Reducing soluble Aß levels and consequently amyloid plaques constitute an attractive therapeutic avenue to, at least, stabilize AD pathogenesis. The brain possesses several mechanisms involved in controlling cerebral Aß levels, among which are the tissue-plasminogen activator (t-PA)/plasmin system and microglia. However, these mechanisms are impaired and ineffective in AD. Here we show that the systemic chronic administration of recombinant t-PA (Activase rt-PA) attenuates AD-related pathology in APPswe/PS1 transgenic mice by reducing cerebral Aß levels and improving the cognitive function of treated mice. Interestingly, these effects do not appear to be mediated by rt-PA-induced plasmin and matrix metalloproteinases 2/9 activation. We observed that rt-PA essentially mediated a slight transient increase in the frequency of patrolling monocytes in the circulation and stimulated microglia in the brain to adopt a neuroprotective phenotype, both of which contribute to Aß elimination. Our study unraveled a new role of rt-PA in maintaining the phagocytic capacity of microglia without exacerbating the inflammatory response and therefore might constitute an interesting approach to stimulate the key populations of cells involved in Aß clearance from the brain.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Microglia/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/administração & dosagem , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/genética , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microglia/fisiologia , Monócitos/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Presenilina-1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...