Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Glia ; 65(7): 1072-1088, 2017 07.
Article in English | MEDLINE | ID: mdl-28417486

ABSTRACT

Microglia, the immune cells of the central nervous system, take part in brain development and homeostasis. They derive from primitive myeloid progenitors that originate in the yolk sac and colonize the brain mainly through intensive migration. During development, microglial migration speed declines which suggests that their interaction with the microenvironment changes. However, the matrix-cell interactions allowing dispersion within the parenchyma are unknown. Therefore, we aimed to better characterize the migration behavior and to assess the role of matrix-integrin interactions during microglial migration in the embryonic brain ex vivo. We focused on microglia-fibronectin interactions mediated through the fibronectin receptor α5ß1 integrin because in vitro work indirectly suggested a role for this ligand-receptor pair. Using 2-photon time-lapse microscopy on acute ex vivo embryonic brain slices, we found that migration occurs in a saltatory pattern and is developmentally regulated. Most importantly, there is an age-specific function of the α5ß1 integrin during microglial cortex colonization. At embryonic day (E) 13.5, α5ß1 facilitates migration while from E15.5, it inhibits migration. These results indicate a developmentally regulated function of α5ß1 integrin in microglial migration during colonization of the embryonic brain.


Subject(s)
Aging , Cell Movement/physiology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Gene Expression Regulation, Developmental/genetics , Integrin alpha5beta1/metabolism , Microglia/physiology , Animals , Blood Vessels/physiology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Embryo, Mammalian , Extracellular Matrix/metabolism , Fibronectins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lectins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phycoerythrin/metabolism , Signal Transduction/physiology
2.
Front Cell Neurosci ; 9: 301, 2015.
Article in English | MEDLINE | ID: mdl-26300736

ABSTRACT

Several studies have indicated that inflammation during pregnancy increases the risk for the development of neuropsychiatric disorders in the offspring. Morphological brain abnormalities combined with deviations in the inflammatory status of the brain can be observed in patients of both autism and schizophrenia. It was shown that acute infection can induce changes in maternal cytokine levels which in turn are suggested to affect fetal brain development and increase the risk on the development of neuropsychiatric disorders in the offspring. Animal models of maternal immune activation reproduce the etiology of neurodevelopmental disorders such as schizophrenia and autism. In this study the poly (I:C) model was used to mimic viral immune activation in pregnant mice in order to assess the activation status of fetal microglia in these developmental disorders. Because microglia are the resident immune cells of the brain they were expected to be activated due to the inflammatory stimulus. Microglial cell density and activation level in the fetal cortex and hippocampus were determined. Despite the presence of a systemic inflammation in the pregnant mice, there was no significant difference in fetal microglial cell density or immunohistochemically determined activation level between the control and inflammation group. These data indicate that activation of the fetal microglial cells is not likely to be responsible for the inflammation induced deficits in the offspring in this model.

3.
Histochem Cell Biol ; 142(1): 79-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24553792

ABSTRACT

In this study, the effect of glycine receptor (GlyR) α3 alternative RNA splicing on the distribution of receptors in the membrane of human embryonic kidney 293 cells is investigated using optical super-resolution microscopy. Direct stochastic optical reconstruction microscopy is used to image both α3K and α3L splice variants individually and together using single- and dual-color imaging. Pair correlation analysis is used to extract quantitative measures from the resulting images. Autocorrelation analysis of the individually expressed variants reveals clustering of both variants, yet with differing properties. The cluster size is increased for α3L compared to α3K (mean radius 92 ± 4 and 56 ± 3 nm, respectively), yet an even bigger difference is found in the cluster density (9,870 ± 1,433 and 1,747 ± 200 µm(-2), respectively). Furthermore, cross-correlation analysis revealed that upon co-expression, clusters colocalize on the same spatial scales as for individually expressed receptors (mean co-cluster radius 94 ± 6 nm). These results demonstrate that RNA splicing determines GlyR α3 membrane distribution, which has consequences for neuronal GlyR physiology and function.


Subject(s)
Cell Membrane/metabolism , Receptors, Glycine/analysis , Cells, Cultured , HEK293 Cells , Humans , Microscopy, Fluorescence , RNA Splicing/genetics , Receptors, Glycine/genetics , Receptors, Glycine/metabolism
4.
Glia ; 61(2): 150-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23001583

ABSTRACT

Microglia are the immune cells of the central nervous system. They are suspected to play important roles in adult synaptogenesis and in the development of the neuronal network. Microglial cells originate from progenitors in the yolk sac. Although it was suggested that they invade the cortex at early developmental stages in the embryo, their invasion pattern remains largely unknown. To address this issue we analyzed the pattern of cortical invasion by microglial cells in mouse embryos at the onset of neuronal cell migration using in vivo immunohistochemistry and ex vivo time-lapse analysis of microglial cells. Microglial cells begin to invade the cortex at 11.5 days of embryonic age (E11.5). They first accumulate at the pial surface and within the lateral ventricles, after which they spread throughout the cortical wall, avoiding the cortical plate region in later embryonic ages. The invasion of the cortical parenchyma occurs in different phases. First, there is a gradual increase of microglial cells between E10.5 and E14.5. From E14.5 to E15.5 there is a rapid phase with a massive increase in microglia, followed by a slow phase again from E15.5 until E17.5. At early stages, many peripheral microglia are actively proliferating before entering the parenchyma. Remarkably, activated microglia accumulate in the choroid plexus primordium, where they are in the proximity of dying cells. Time-lapse analysis shows that embryonic microglia are highly dynamic cells.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/embryology , Embryonic Development/physiology , Microglia/physiology , Age Factors , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Cell Movement , Cell Proliferation , Choroid Plexus/cytology , Choroid Plexus/embryology , Embryo, Mammalian/anatomy & histology , Female , Galectin 3/metabolism , Green Fluorescent Proteins/genetics , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Confocal , Neurons/physiology , Pregnancy , Receptors, Interleukin-8A/genetics
5.
J Neurosci ; 32(34): 11559-73, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915101

ABSTRACT

Microglia are known to invade the mammalian spinal cord (SC) at an early embryonic stage. While the mechanisms underlying this early colonization of the nervous system are still unknown, we recently found that it is associated, at least partially, with the ability of microglia to proliferate at the onset of motoneuron developmental cell death and of synaptogenesis in mouse embryo (E13.5). In vitro studies have shown that the proliferation and activation of adult microglia can be influenced by the purinergic ionotropic receptor P2X7 via a coupling with Pannexin-1. By performing patch-clamp recordings in situ using a whole-mouse embryonic SC preparation, we show here that embryonic microglia already express functional P2X7R. P2X7R activation evoked a biphasic current in embryonic microglia, which is supposed to reflect large plasma membrane pore opening. However, although embryonic microglia express pannexin-1, this biphasic current was still recorded in microglia of pannexin-1 knock-out embryos, indicating that it rather reflected P2X7R intrinsic pore dilatation. More important, we found that proliferation of embryonic SC microglia, but not their activation state, depends almost entirely on P2X7R by comparing wild-type and P2X7R-/- embryos. Absence of P2X7R led also to a decrease in microglia density. Pannexin-1-/- embryos did not exhibit any difference in microglial proliferation, showing that the control of embryonic microglial proliferation by P2X7R does not depend on pannexin-1 expression. These results reveal a developmental role of P2X7R by controlling embryonic SC microglia proliferation at a critical developmental state in the SC of mouse embryos.


Subject(s)
Cell Differentiation/physiology , Connexins/metabolism , Microglia/physiology , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , Spinal Cord/cytology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Antigens, CD/metabolism , Biophysics , CX3C Chemokine Receptor 1 , Caspase 3/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Connexins/deficiency , Electric Stimulation , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Ki-67 Antigen/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Patch-Clamp Techniques , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , RNA, Messenger/metabolism , Receptors, Chemokine/genetics , Receptors, Purinergic P2X7/deficiency , Receptors, Purinergic P2X7/genetics , Rosaniline Dyes , Spinal Cord/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL