Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 17: 1120400, 2023.
Article in English | MEDLINE | ID: mdl-37006469

ABSTRACT

During development microglia colonize the central nervous system (CNS) and play an important role in programmed cell death, not only because of their ability to remove dead cells by phagocytosis, but also because they can promote the death of neuronal and glial cells. To study this process, we used as experimental systems the developing in situ quail embryo retina and organotypic cultures of quail embryo retina explants (QEREs). In both systems, immature microglia show an upregulation of certain inflammatory markers, e.g., inducible NO synthase (iNOS), and nitric oxide (NO) under basal conditions, which can be further enhanced with LPS-treatment. Hence, we investigated in the present study the role of microglia in promoting ganglion cell death during retinal development in QEREs. Results showed that LPS-stimulation of microglia in QEREs increases (i) the percentage of retinal cells with externalized phosphatidylserine, (ii) the frequency of phagocytic contacts between microglial and caspase-3-positive ganglion cells, (iii) cell death in the ganglion cell layer, and (iv) microglial production of reactive oxygen/nitrogen species, such as NO. Furthermore, iNOS inhibition by L-NMMA decreases cell death of ganglion cells and increases the number of ganglion cells in LPS-treated QEREs. These data demonstrate that LPS-stimulated microglia induce ganglion cell death in cultured QEREs by a NO-dependent mechanism. The fact that phagocytic contacts between microglial and caspase-3-positive ganglion cells increase suggests that this cell death might be mediated by microglial engulfment, although a phagocytosis-independent mechanism cannot be excluded.

2.
Front Cell Neurosci ; 16: 816439, 2022.
Article in English | MEDLINE | ID: mdl-35197828

ABSTRACT

Microglia are the tissue-resident macrophages of the central nervous parenchyma. In mammals, microglia are thought to originate from yolk sac precursors and posteriorly maintained through the entire life of the organism. However, the contribution of microglial cells from other sources should also be considered. In addition to "true" or "bona-fide" microglia, which are of embryonic origin, the so-called "microglia-like cells" are hematopoietic cells of bone marrow origin that can engraft the mature brain mainly under pathological conditions. These cells implement great parts of the microglial immune phenotype, but they do not completely adopt the "true microglia" features. Because of their pronounced similarity, true microglia and microglia-like cells are usually considered together as one population. In this review, we discuss the origin and development of these two distinct cell types and their differences. We will also review the factors determining the appearance and presence of microglia-like cells, which can vary among species. This knowledge might contribute to the development of therapeutic strategies aiming at microglial cells for the treatment of diseases in which they are involved, for example neurodegenerative disorders like Alzheimer's and Parkinson's diseases.

3.
Glia ; 69(4): 842-857, 2021 04.
Article in English | MEDLINE | ID: mdl-33105046

ABSTRACT

Activation of microglia is an early immune response to damage in the brain. Although a key role for Ca2+ as trigger of microglial activation has been considered, little is known about the molecular scenario for regulating Ca2+ homeostasis in these cells. Taking into account the importance of the endoplasmic reticulum as a cellular Ca2+ store, the sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA2b) is an interesting target to modulate intracellular Ca2+ dynamics. We found upregulation of SERCA2b in activated microglia of human brain with Alzheimer's disease and we further studied the participation of SERCA2b in microglial functions by using the BV2 murine microglial cell line and primary microglia isolated from mouse brain. To trigger microglia activation, we used the bacterial lipopolysaccharide (LPS), which is known to induce an increase of cytosolic Ca2+ . Our results showed an upregulated expression of SERCA2b in LPS-induced activated microglia likely associated to an attempt to restore the increased cytosolic Ca2+ concentration. We analyzed SERCA2b contribution in microglial migration by using the specific SERCA inhibitor thapsigargin in scratch assays. Microglial migration was strongly stimulated with thapsigargin, even more than with LPS-induction, but delayed in time. However, phagocytic capacity of microglia was blocked in the presence of the SERCA inhibitor, indicating the importance of a tight control of cytosolic Ca2+ in these processes. All together, these results provide for the first time compelling evidence for SERCA2b as a major player regulating microglial functions, affecting migration and phagocytosis in an opposite manner.


Subject(s)
Microglia , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Lipopolysaccharides/toxicity , Mice , Microglia/metabolism , Phagocytosis , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/pharmacology
4.
PLoS One ; 12(8): e0182450, 2017.
Article in English | MEDLINE | ID: mdl-28763502

ABSTRACT

Microglial cell precursors located in the area of the base of the pecten and the optic nerve head (BP/ONH) start to enter the retina of quail embryos at the 7th day of incubation (E7), subsequently colonizing the entire retina by central-to-peripheral tangential migration, as previously shown by our group. The present study demonstrates a precise chronological coincidence of the onset of microglial cell entry into the retina with a striking increase in death of retinal cells, as revealed by their active caspase-3 expression and TUNEL staining, in regions dorsal to the BP/ONH area, suggesting that dying retinal cells would contribute to the microglial cell inflow into the retina. However, the molecular mechanisms involved in this inflow are currently unclear. Extracellular nucleotides, such as ATP and UDP, have previously been shown to favor migration of microglia towards brain injuries because they are released by apoptotic cells and stimulate both chemotaxis and chemokinesis in microglial cells via signaling through purinergic receptors. Hence, we tested here the hypothesis that ATP and UDP play a role in the entry and migration of microglial precursors into the developing retina. For this purpose, we used an experimental model system based on organotypic cultures of E6.5 quail embryo retina explants, which mimics the entry and migration of microglial precursors in the in situ developing retina. Inhibition of purinergic signaling by treating retina explants with either apyrase, a nucleotide-hydrolyzing enzyme, or suramin, a broad spectrum antagonist of purinergic receptors, significantly prevents the entry of microglial cells into the retina. In addition, treatment of retina explants with either exogenous ATP or UDP results in significantly increased numbers of microglial cells entering the retina. In light of these findings, we conclude that purinergic signaling by extracellular ATP and UDP is necessary for the entry and migration of microglial cells into the embryonic retina by inducing chemokinesis in these cells.


Subject(s)
Adenosine Triphosphate/metabolism , Caspase 3/metabolism , Gene Expression Regulation, Developmental , Microglia/cytology , Retina/embryology , Uridine Diphosphate/metabolism , Animals , Cell Survival , Chemotaxis , Enzyme Activation , Microscopy, Confocal , Optic Nerve/pathology , Quail , Receptors, Purinergic/metabolism , Retina/physiology , Signal Transduction , Time Factors
5.
PLoS One ; 10(8): e0135238, 2015.
Article in English | MEDLINE | ID: mdl-26252475

ABSTRACT

The role of microglia during neurodegeneration remains controversial. We investigated whether microglial cells have a neurotoxic or neuroprotective function in the retina. Retinal explants from 10-day-old mice were treated in vitro with minocycline to inhibit microglial activation, with LPS to increase microglial activation, or with liposomes loaded with clodronate (Lip-Clo) to deplete microglial cells. Flow cytometry was used to assess the viability of retinal cells in the explants and the TUNEL method to show the distribution of dead cells. The immunophenotypic and morphological features of microglia and their distribution were analyzed with flow cytometry and immunocytochemistry. Treatment of retinal explants with minocycline reduced microglial activation and simultaneously significantly decreased cell viability and increased the presence of TUNEL-labeled cell profiles. This treatment also prevented the migration of microglial cells towards the outer nuclear layer, where cell death was most abundant. The LPS treatment increased microglial activation but had no effect on cell viability or microglial distribution. Finally, partial microglial removal with Lip-Clo diminished the cell viability in the retinal explants, showing a similar effect to that of minocycline. Hence, cell viability is diminished in retinal explants cultured in vitro when microglial cells are removed or their activation is inhibited, indicating a neurotrophic role for microglia in this system.


Subject(s)
Clodronic Acid/chemistry , Microglia/cytology , Optic Nerve/growth & development , Retina/growth & development , Animals , Animals, Newborn , Cell Survival , Clodronic Acid/administration & dosage , Escherichia coli , Flow Cytometry , Immunohistochemistry , Immunophenotyping , Lipopolysaccharides/chemistry , Liposomes/chemistry , Mice , Mice, Inbred C57BL , Minocycline/chemistry , Neuroprotection , Optic Nerve/drug effects , Organ Culture Techniques , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Retina/cytology , Retina/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Invest Ophthalmol Vis Sci ; 56(2): 1301-9, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25650421

ABSTRACT

PURPOSE: The purpose of this study was to investigate the incidence of DNA damage during postnatal development of the retina and the relationship between DNA damage and cell death. METHODS: DNA damage in the developing postnatal retina of C57BL/6 mice was assessed by determining the amounts of 8-hydroxy-2'-deoxyguanosine (8-OHdG), which is indicative of DNA oxidation and related to the formation of DNA single-strand breaks (SSBs), and phosphorylated histone H2AX (γ-H2AX), a marker of DNA double-strand breaks (DSBs). Poly(ADP-ribose) polymerase (PARP) activation was measured by ELISA and Western blotting. The location of γ-H2AX-positive and dying cells was determined by immunofluorescence and TUNEL assays. RESULTS: Oxidative DNA damage was maintained at low levels during high PARP activation between postnatal days 0 (P0) and P7. Phosphorylated histone H2AX gradually increased between P0 and P14 and decreased thereafter. Phosphorylated histone H2AX-positive cells with cell death morphology or TUNEL positivity were more abundant at P7 than at P14. CONCLUSIONS: Oxidative DNA damage in postnatal retina increases during development. It is low during the first postnatal week when PARP-1 activity is high but increases thereafter. The rise in DSBs when PARP activity is downregulated may be attributable to accumulated oxidative damage and SSBs. At P7 and P14, γ-H2AX-positive cells are repairing naturally occurring DNA damage, but some are dying (mostly at P7), probably due to an accumulation of irreparable DNA damage.


Subject(s)
DNA Damage/genetics , DNA/genetics , Gene Expression Regulation, Developmental , Histones/genetics , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/genetics , Retina/growth & development , Animals , Animals, Newborn , Apoptosis , Blotting, Western , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Histones/biosynthesis , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/biosynthesis
7.
PLoS One ; 9(8): e106048, 2014.
Article in English | MEDLINE | ID: mdl-25170849

ABSTRACT

Inducible nitric oxide synthase (iNOS), which produce large amounts of nitric oxide (NO), is induced in macrophages and microglia in response to inflammatory mediators such as LPS and cytokines. Although iNOS is mainly expressed by microglia that become activated in different pathological and experimental situations, it was recently reported that undifferentiated amoeboid microglia can also express iNOS during normal development. The aim of this study was to investigate the pattern of iNOS expression in microglial cells during normal development and after their activation with LPS by using the quail retina as model. iNOS expression was analyzed by iNOS immunolabeling, western-blot, and RT-PCR. NO production was determined by using DAR-4M AM, a reliable fluorescent indicator of subcellular NO production by iNOS. Embryonic, postnatal, and adult in situ quail retinas were used to analyze the pattern of iNOS expression in microglial cells during normal development. iNOS expression and NO production in LPS-treated microglial cells were investigated by an in vitro approach based on organotypic cultures of E8 retinas, in which microglial cell behavior is similar to that of the in situ retina, as previously demonstrated in our laboratory. We show here that amoeboid microglia in the quail retina express iNOS during normal development. This expression is stronger in microglial cells migrating tangentially in the vitreal part of the retina and is downregulated, albeit maintained, when microglia differentiate and become ramified. LPS treatment of retina explants also induces changes in the morphology of amoeboid microglia compatible with their activation, increasing their lysosomal compartment and upregulating iNOS expression with a concomitant production of NO. Taken together, our findings demonstrate that immature microglial cells express iNOS during normal development, suggesting a certain degree of activation. Furthermore, LPS treatment induces overactivation of amoeboid microglia, resulting in a significant iNOS upregulation.


Subject(s)
Avian Proteins/metabolism , Microglia/enzymology , Nitric Oxide Synthase Type II/metabolism , Retina/enzymology , Animals , Animals, Newborn , Avian Proteins/genetics , Blotting, Western , Coturnix , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microscopy, Confocal , Nitric Oxide Synthase Type II/genetics , Retina/embryology , Retina/growth & development , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques
8.
Exp Eye Res ; 121: 42-57, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24582572

ABSTRACT

Organotypic cultures of retinal explants allow the detailed analysis of microglial cells in a cellular microenvironment similar to that in the in situ retina, with the advantage of easy experimental manipulation. However, the in vitro culture causes changes in the retinal cytoarchitecture and induces a microglial response that may influence the results of these manipulations. The purpose of this study was to analyze the influence of the retinal age on changes in retinal cytoarchitecture, cell viability and death, and microglial phenotype and distribution throughout the in vitro culture of developing and adult retina explants. Explants from developing (3 and 10 postnatal days, P3 and P10) and adult (P60) mouse retinas were cultured for up to 10 days in vitro (div). Dead or dying cells were recognized by TUNEL staining, cell viability was determined by flow cytometry, and the numbers and distribution patterns of microglial cells were studied by flow cytometry and immunocytochemistry, respectively. The retinal cytoarchitecture was better preserved at prolonged culture times (10 div) in P10 retina explants than in P3 or adult explants. Particular patterns of cell viability and death were observed at each age: in general, explants from developing retinas showed higher cell viability and lower density of TUNEL-positive profiles versus adult retinas. The proportion of microglial cells relative to the whole population of retinal cells was higher in explants fixed immediately after their dissection (i.e., non-cultured) from adult retinas than in those from developing retinas. This proportion was always higher in non-cultured explants than in explants at 10 div, suggesting the death of some microglial cells during the culture. Activation of microglial cells, as revealed by their phenotypical appearance, was observed in both developing and adult retina explants from the beginning of the culture. Immunofluorescence with the anti-CD68 antibody showed that some activated microglial cells were CD68-positive but others were CD68-negative. Flow cytometry using CD68-labeling revealed that the percentage of CD68-positive microglial cells was much higher in developing than in adult retina explants, despite the activation of microglia in both types of explants, indicating that CD68-labeling was more closely related to the maturity degree of microglia than to their activation. Some swollen activated microglial cells entered the outer nuclear layer in developing and adult cultured retinal explants, whereas this layer was devoid of microglia in non-cultured explants. There was no apparent correlation between the distribution of microglia and that of TUNEL-labeled profiles. However, some swollen activated microglial cells in the outer and inner nuclear layers engulfed clusters of cell nuclei that were negative or weakly positive for TUNEL. This engulfment activity of microglia mimicked that observed in degenerative pathologies of the retina. We conclude that organotypic cultures from developing retinas show a higher rate of cell viability and better preservation of the normal cytoarchitecture in comparison to those obtained from adult retinas. In addition, the features of microglial response in cultured retinal explants show them to be a useful model for studying interactions between microglial cells and degenerating neurons in retinal diseases.


Subject(s)
Aging/physiology , Microglia/cytology , Retina/growth & development , Animals , Animals, Newborn , CD11b Antigen/metabolism , Cell Death/physiology , Cell Survival/physiology , Flow Cytometry , In Situ Nick-End Labeling , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Organ Culture Techniques , Retina/metabolism
9.
Invest Ophthalmol Vis Sci ; 52(10): 7445-54, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21705688

ABSTRACT

PURPOSE: Poly(ADP-ribose) polymerase (PARP)-1 is a nuclear enzyme that transfers ADP-ribose units (PAR polymer) to nuclear proteins and has been implicated in caspase-independent cell death in different models of retinal degeneration. The involvement of PARP-1 in cell death occurring during normal postnatal development of the mouse retina was investigated. In addition, the expression of apoptosis-inducing factor (AIF), a caspase-independent cell death mediator, was explored because PARP-1 activation has been related to the translocation of a 57-kDa form of AIF into the cell nucleus. METHODS: Cell death was determined in retinas of developing mice by both ELISA and TUNEL. PARP-1, PAR, and AIF were analyzed by immunocytochemistry and immunoblotting. Quantification of PARP-1 mRNA levels was also performed by real-time PCR. RESULTS: PARP-1 upregulation and PAR polymer formation, indicative of PARP-1 activity, were observed during the first postnatal week simultaneously with the presence of abundant dying cells, some of which were not associated with active caspase-3. PARP-1 was downregulated and PARP-1 activity progressively declined in the retina during subsequent postnatal development, coinciding with the decrease in cell death. Truncated AIF (57 kDa) was present in the retina during the first postnatal week, gradually decreasing thereafter, and had a nuclear localization in some cells, which also showed strong PAR polymer nuclear staining. CONCLUSIONS: These results show that a caspase-independent cell death pathway exists during the normal development of the mouse retina and suggest that PARP-1 participates in this cell death pathway by mediating AIF translocation to the cell nucleus.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Enzymologic/physiology , Poly(ADP-ribose) Polymerases/genetics , Retina/enzymology , Retina/growth & development , Animals , Animals, Newborn , Apoptosis Inducing Factor/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nucleosomes , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Dev Neurobiol ; 71(4): 296-315, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21394933

ABSTRACT

Organotypic cultures of retina explants preserve the complex cellular microenvironment of the retina and have been used as a tool to assess the biological functions of some cell types. However, studies to date have shown that microglial cells activate quickly in response to the retina explantation. In this study, microglial cells migrated and ramified in quail embryo retina organotypic cultures (QEROCs) according to chronological patterns bearing a resemblance to those in the retina in situ, despite some differences in cell density and ramification degree. Retinal explants from quail embryos at 9 days of incubation (E9) proved to be the best in vitro system for reproducing a physiological-like behavior of microglial cells when cultured in Eagle's basal medium supplemented with horse serum. During the first week in vitro, microglial cells migrated tangentially in the vitreal part of QEROCs, and some began to migrate radially from 3 days in vitro (div) onward, ramifying in the inner and outer plexiform layers, thus mimicking microglia development in the retina in situ, although reaching a lower degree of ramification after 7 div. From 8 div onward, microglial cells rounded throughout the explant thickness simultaneously with the nonphysiological appearance of dead photoreceptors and round microglia in the outernuclear layer. Therefore, E9 QEROCs can be used during the first week in vitro as a model system for experimental studies of molecules putatively involved in microglial migration and ramification.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Microglia/physiology , Retina/cytology , Retina/embryology , Animals , Cell Culture Techniques , Coturnix , Microglia/cytology , Organ Culture Techniques , Photoreceptor Cells/cytology , Photoreceptor Cells/physiology
11.
Neuron Glia Biol ; 7(1): 25-40, 2011 Feb.
Article in English | MEDLINE | ID: mdl-22377033

ABSTRACT

Microglia, the brain's innate immune cell type, are cells of mesodermal origin that populate the central nervous system (CNS) during development. Undifferentiated microglia, also called ameboid microglia, have the ability to proliferate, phagocytose apoptotic cells and migrate long distances toward their final destinations throughout all CNS regions, where they acquire a mature ramified morphological phenotype. Recent studies indicate that ameboid microglial cells not only have a scavenger role during development but can also promote the death of some neuronal populations. In the mature CNS, adult microglia have highly motile processes to scan their territorial domains, and they display a panoply of effects on neurons that range from sustaining their survival and differentiation contributing to their elimination. Hence, the fine tuning of these effects results in protection of the nervous tissue, whereas perturbations in the microglial response, such as the exacerbation of microglial activation or lack of microglial response, generate adverse situations for the organization and function of the CNS. This review discusses some aspects of the relationship between microglial cells and neuronal death/survival both during normal development and during the response to injury in adulthood.


Subject(s)
Apoptosis/physiology , Brain/cytology , Microglia/physiology , Neurons/physiology , Animals , Humans , Macrophages/physiology , Phagocytes/physiology
12.
J Comp Neurol ; 518(4): 477-92, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20020538

ABSTRACT

The microglial response elicited by degeneration of retinal photoreceptor cells was characterized in BALB/c mice exposed to bright light for 7 hours and then kept in complete darkness for survival times ranging from 0 hours to 10 days. Photodegeneration resulted in extensive cell death in the retina, mainly in the outer nuclear layer (ONL), where the photoreceptor nuclei are located. Specific immunolabeling of microglial cells with anti-CD11b, anti-CD45, anti-F4/80, anti-SRA, and anti-CD68 antibodies revealed that microglial cells were activated in light-exposed retinas. They migrated to the ONL, changed their morphology, becoming rounded cells with short and thick processes, and, finally, showed immunophenotypic changes. Specifically, retinal microglia began to strongly express antigens recognized by anti-CD11b, anti-CD45, and anti-F4/80, coincident with cell degeneration. In contrast, upregulation of the antigen recognized by anti-SRA was not detected by immunocytochemistry until 6 hours after light exposure. Differences were also observed at 10 days after light exposure: CD11b, CD45, and F4/80 continued to be strongly expressed in retinal microglia, whereas the expression of CD68 and SRA had decreased to near-normal values. Therefore, microglia did not return to their original state after photodegeneration and continued to show a degree of activation. The accumulation of activated microglial cells in affected regions simultaneously with photoreceptor degeneration suggests that they play some role in photodegeneration.


Subject(s)
Gliosis/physiopathology , Light/adverse effects , Microglia/physiology , Microglia/radiation effects , Retinal Degeneration/physiopathology , Animals , Antibody Specificity/immunology , Antigens, Surface/analysis , Antigens, Surface/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Cell Movement/immunology , Cell Shape , Chemotaxis/immunology , Darkness , Disease Models, Animal , Gliosis/etiology , Gliosis/pathology , Immunohistochemistry , Immunophenotyping , Male , Mice , Mice, Inbred BALB C , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Retinal Degeneration/etiology , Retinal Degeneration/pathology
13.
J Comp Neurol ; 506(2): 224-39, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18022954

ABSTRACT

Macrophage/microglial cells in the mouse retina during embryonic and postnatal development were studied by immunocytochemistry with Iba1, F4/80, anti-CD45, and anti-CD68 antibodies and by tomato lectin histochemistry. These cells were already present in the retina of embryos aged 11.5 days (E11.5) in association with cell death. At E12.5 some macrophage/microglial cells also appeared in peripheral regions of the retina with no apparent relationship with cell death. Immediately before birth microglial cells were present in the neuroblastic, inner plexiform (IPL), and ganglion cell (GCL) layers, and their distribution suggested that they entered the retina from the ciliary margin and the vitreous. The density of retinal microglial cells strongly decreased at birth, increased during the first postnatal week as a consequence of the entry of microglial precursors into the retina from the vitreous, and subsequently decreased owing to the cessation of microglial entry and the increase in retina size. The mature topographical distribution pattern of microglia emerged during postnatal development of the retina, apparently by radial migration of microglial cells from the vitreal surface in a vitreal-to-scleral direction. Whereas microglial cells were only seen in the GCL and IPL at birth, they progressively appeared in more scleral layers at increasing postnatal ages. Thus, microglial cells were present within all layers of the retina except the outer nuclear layer at the beginning of the second postnatal week. Once microglial cells reached their definitive location, they progressively ramified.


Subject(s)
Microglia/physiology , Retina , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Calcium-Binding Proteins/metabolism , Cell Count , Cell Differentiation , Embryo, Mammalian , In Situ Nick-End Labeling , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microfilament Proteins , Plant Lectins/pharmacokinetics , Retina/cytology , Retina/embryology , Retina/growth & development
14.
Glia ; 54(5): 376-93, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16886202

ABSTRACT

Ameboid microglial cells migrate tangentially on the vitreal part of quail embryo retinas by crawling on Müller cell end-feet (MCEF) to which they adhere. These microglial cells can be cultured immediately after dissection of the eye and isolation of sheets containing the inner limiting membrane (ILM) covered by a carpet of MCEF (ILM/MCEF sheets), to which the cells remain adhered. Morphological changes of microglial cells cultured on ILM/MCEF sheets for 4 days were characterized in this study. During the first minutes in vitro, lamellipodia-bearing bipolar microglial cells became rounded in shape. From 1 to 24 h in vitro (hiv), microglial cells swept and phagocytosed the MCEF on which they were initially adhered, becoming directly adhered on the ILM. MCEF sweep was dependent on active cell motility, as shown by inhibition of sweep after cytochalasin D treatment. From 24 hiv on, after MCEF phagocytosis, microglial cells became more flattened, increasing the surface area of their adhesion to substrate, and expressed the beta1 subunit of integrins on their membrane. Morphological evidence suggested that microglial cells migrated for short distances on ILM/MCEF sheets, leaving tracks produced by their strong adhesion to the substrate. The simplicity of the isolation method, the immediate availability of cultured microglial cells, and the presence of multiple functional processes (phagocytosis, migration, upregulation of surface molecules, etc.) make cultures of microglial cells on ILM/MCEF sheets a valuable model system for in vitro experimental investigation of microglial cell functions.


Subject(s)
Cell Movement/physiology , Microglia/cytology , Microglia/physiology , Organogenesis/physiology , Retina/cytology , Retina/embryology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Culture Techniques/methods , Cell Movement/drug effects , Cell Polarity/drug effects , Cell Polarity/physiology , Cell Shape/drug effects , Cell Shape/physiology , Cells, Cultured , Coturnix , Cytochalasin D/pharmacology , Fluorescent Antibody Technique , Microglia/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Organogenesis/drug effects , Phagocytosis/drug effects , Phagocytosis/physiology , Pseudopodia/drug effects , Pseudopodia/physiology , Pseudopodia/ultrastructure
15.
J Comp Neurol ; 492(1): 20-33, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16175556

ABSTRACT

Activation of mature (ramified) microglia in response to injury in the adult central nervous system (CNS) is well documented. However, the response of immature (ameboid) microglia to injury in the developing CNS has received little attention. In this study, a stab wound was made in embryonic quail retinas at incubation days 7 and 9, and the response of retinal microglial cells was analyzed at different times between days 1 and 37 postinjury. The appearance of microglial cells within the wound occurred at the same time as the arrival of the first migrating ameboid microglial cells at an equivalent area in control retinas. Therefore, no specific attraction of microglia toward the wound was observed. Microglial cells in the wound had phenotypic features similar to those of activated microglia in the adult CNS. Thus, their shape was more compact compared with microglial cells outside the wound, expression of the molecule recognized by the QH1 antibody was up-regulated, and their lysosomal compartment was markedly increased. Transitional forms between normal ameboid and activated-like microglial cells were seen at the wound edge, supporting the view that ameboid microglia become activated when they contact the wound during the normal course of their migration in the retina. The microglial reaction was maintained within the wound at 37 days postinjury. In addition to the stab wound, secondary damage areas were found in experimental retinas. Activated cells could still be observed in these areas at 37 days postinjury.


Subject(s)
Coturnix/embryology , Microglia/cytology , Microglia/physiology , Retina/cytology , Retina/embryology , Wound Healing/physiology , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Cell Shape/physiology , Embryo, Nonmammalian , Microglia/pathology , Retina/injuries , Wounds, Stab/pathology , Wounds, Stab/physiopathology
16.
Glia ; 46(3): 261-73, 2004 May.
Article in English | MEDLINE | ID: mdl-15048849

ABSTRACT

Microglial cells spread within the nervous system by tangential and radial migration. The cellular mechanism of tangential migration of microglia has been described in the quail retina but the mechanism of their radial migration has not been studied. In this work, we clarify some aspects of this mechanism by analyzing morphological features of microglial cells at different steps of their radial migration in the quail retina. Microglial cells migrate in the vitreal half of the retina by successive jumps from the vitreal border to progressively more scleral levels located at the vitreal border, intermediate regions, and scleral border of the inner plexiform layer (IPL). The cellular mechanism used for each jump consists of the emission of a leading thin radial process that ramifies at a more scleral level before retraction of the rear of the cell. Hence, radial migration and ramification of microglial cells are simultaneous events. Once at the scleral border of the IPL, microglial cells migrate through the inner nuclear layer to the outer plexiform layer by another mechanism: they retract cell processes, become round, and squeeze through neuronal bodies. Microglial cells use radial processes of s-laminin-expressing Müller cells as substratum for radial migration. Levels where microglial cells stop and ramify at each jump are always interfaces between retinal strata with strong tenascin immunostaining and strata showing weak or no tenascin immunoreactivity. When microglial cell radial migration ends, tenascin immunostaining is no longer present in the retina. These findings suggest that tenascin plays a role in the stopping and ramification of radially migrating microglial cells.


Subject(s)
Cell Movement/physiology , Coturnix/physiology , Microglia/cytology , Retina/cytology , Animals , Animals, Newborn , Cell Differentiation/physiology , Coturnix/embryology , Coturnix/growth & development , Microglia/physiology , Microscopy, Confocal/methods , Retina/embryology , Retina/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...