13 research outputs found
Implication du remodelage de l'unité neurovasculaire dans la maladie d'Alzheimer : l'hypoperfusion cérébrale et le systÚme de l'activateur tissulaire du plasminogÚne
Tableau dâhonneur de la FacultĂ© des Ă©tudes supĂ©rieures et postdoctorales, 2015-2016LâunitĂ© neurovasculaire (NVU) est centrale dans lâĂ©limination de la ÎČ-amyloĂŻde dont lâaccumulation promeut le dĂ©veloppement de la maladie dâAlzheimer (AD). Suivant une perturbation vasculaire, le bris ou lâaltĂ©ration de la barriĂšre hĂ©matoencĂ©phalique induit le remodelage de la NVU. Par exemple, les cellules endothĂ©liales sĂ©crĂštent lâactivateur tissulaire du plasminogĂšne (t-PA), ce qui module les cellules composant la NVU. Câest pourquoi, nous nous sommes intĂ©ressĂ©s Ă ce remodelage dans la AD en Ă©tudiant lâeffet de lâhypoperfusion cĂ©rĂ©brale chronique sĂ©vĂšre (SCCH) et de lâadministration du t-PA. Suite Ă la SCCH, les souris dĂ©veloppant la AD, APPswe/PS1, dĂ©montrent un dĂ©clin cognitif plus important causĂ© par un dysfonctionnement des microglies. En contre partie, nous avons observĂ© une amĂ©lioration des fonctions cognitives des APPswe/PS1 suite Ă lâinjection systĂ©mique du t-PA qui induit lâactivation des microglies via la protĂ©ine apparentĂ©e au rĂ©cepteur des protĂ©ines de faibles densitĂ©, LRP1, et promeut lâĂ©limination de lâAÎČ. Ainsi, nos rĂ©sultats dĂ©montrent que le remodelage de la NVU peut aggraver la pathogenĂšse, mais Ă©galement fournir des pistes de traitement.Brain remodeling by the neurovascular unit (NVU) has gain interest in disease such as Alzheimerâs disease (AD). Following vascular perturbation, NVU go through remodeling due to disruption or alteration of brain-blood barrier. One of the molecule inducing remodeling is the tissue-plasminogen activator (t-PA) released by endothelial cells. In fact, t-PA can act both as an enzyme and a cytokine. Thus, we studied the effect of vascular perturbation and t-PA system in AD. By developing a new model of a severe chronic cerebral hypoperfusion (SCCH), we demonstrate that SCCH aggravates memory loss in AD mice, APPswe/PS1, due to microglia dysfunction. Indeed, low glucose environment lowers microgliaâs activity and phagocytosis capacity. On the other hand, systemic administration of t-PA improves cognition as well as decreases amyloid burden in APPswe/PS1. Acting as a cytokine, rt-PA binds LRP1 which induces microgliaâs activation and promotes amyloid elimination. These data suggest that NVU remodeling occurring in AD may participate in the disease pathogenesis and provide new insight of treatment, such as rt-PA
Neuropathobiology of COVID-19 : The Role for Glia
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.Medicine, Faculty ofNon UBCBiochemistry and Molecular Biology, Department ofReviewedFacultyResearche
Microglial and peripheral immune priming is partially sexually dimorphic in adolescent mouse offspring exposed to maternal high-fat diet
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 macrophagesâmicroglia. 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.Medicine, Faculty ofNon UBCBiochemistry and Molecular Biology, Department ofReviewedFacult
Single-cell transcriptomics of the ventral posterolateral nucleus-enriched thalamic regions from HSV-1-infected mice reveal a novel microglia/microglia-like transcriptional response
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 CD11bâș 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) (6âĂâ10â” PFUs/20 ”l). 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 âin transitionâ 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-1ÎČ production, promoting a pro-inflammatory response. These cells' increased expression of viral transcripts suggests that the distinct âin transitionâ 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 TMEM119âș/IBA-Iâș 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.Medicine, Faculty ofNon UBCBiochemistry and Molecular Biology, Department ofReviewedFacultyResearche
Microglial homeostasis disruption modulates non-rapid eye movement sleep duration and neuronal activity in adult female mice.
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-1ÎČ 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
Microglia are involved in phagocytosis and extracellular digestion during Zika virus encephalitis in young adult immunodeficient mice
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 (1âĂâ10â” PFUs/100 ”L).
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 (1.3âĂâ10â” genome copies/mL and 9.8âĂâ10â· 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 Iba1âș/TMEM119âș 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 (2.8âĂâ10Âčâ°
versus 8.5âĂâ10âž 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.Medicine, Faculty ofNon UBCBiochemistry and Molecular Biology, Department ofReviewedFacult
Maternal high-fat diet modifies myelin organization, microglial interactions, and results in social memory and sensorimotor gating deficits in adolescent mouse offspring
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 âa growth factor released by microgliaâ 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
Levodopa partially rescues microglial numerical, morphological, and phagolysosomal alterations in a monkey model of Parkinsonâs disease
International audienceParkinson'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
Erratum to âN-3 PUFA deficiency disrupts oligodendrocyte maturation and myelin integrity during brain developmentâ
This article corrects the following:N-3 PUFA deficiency disrupts oligodendrocyte maturation and myelin integrity during brain developmentQuentin Leyrolle, Fanny Decoeur, Cyril Dejean, Galadriel BriÚre, Stephane Leon, Ioannis Bakoyiannis, Emilie Baroux, Tony-Lee Sterley, Clémentine Bosch-Bouju, Lydie Morel, Camille Amadieu, Cynthia Lecours, Marie-Kim St-Pierre, Maude Bordeleau, Helene Roumes, Véronique De Smedt-Peyrusse, Alexandran Séré, Leslie Schwendimann, Stephane Grégoire, Lionel Bretillon, Niyazi Acar, Corinne Joffre, Guillaume Ferreira, Raluca Uricaru, Patricia Thebault, Pierre Gressens, Marie-Eve Tremblay, Sophie Layé, Agnes Nadjar Volume 70Issue 1Glia pages: 50-70 First Published online: September 14, 2021International audienc