Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Glia ; 71(2): 415-430, 2023 02.
Article in English | MEDLINE | ID: mdl-36308278

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are uniformly distributed in the mammalian brain; however, their function is rather heterogeneous in respect to their origin, location, receptor/channel expression and age. The basic helix-loop-helix transcription factor Olig2 is expressed in all OPCs as a pivotal determinant of their differentiation. Here, we identified a subset (2%-26%) of OPCs lacking Olig2 in various brain regions including cortex, corpus callosum, CA1 and dentate gyrus. These Olig2 negative (Olig2neg ) OPCs were enriched in the juvenile brain and decreased subsequently with age, being rarely detectable in the adult brain. However, the loss of this population was not due to apoptosis or microglia-dependent phagocytosis. Unlike Olig2pos OPCs, these subset cells were rarely labeled for the mitotic marker Ki67. And, accordingly, BrdU was incorporated only by a three-day long-term labeling but not by a 2-hour short pulse, suggesting these cells do not proliferate any more but were derived from proliferating OPCs. The Olig2neg OPCs exhibited a less complex morphology than Olig2pos ones. Olig2neg OPCs preferentially remain in a precursor stage rather than differentiating into highly branched oligodendrocytes. Changing the adjacent brain environment, for example, by acute injuries or by complex motor learning tasks, stimulated the transition of Olig2pos OPCs to Olig2neg cells in the adult. Taken together, our results demonstrate that OPCs transiently suppress Olig2 upon changes of the brain activity.


Subject(s)
Brain Injuries , Oligodendrocyte Precursor Cells , Animals , Oligodendrocyte Precursor Cells/metabolism , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Brain Injuries/metabolism , Mammals/metabolism
2.
Pflugers Arch ; 475(9): 1035-1044, 2023 09.
Article in English | MEDLINE | ID: mdl-37401986

ABSTRACT

In the central nervous system, oligodendrocyte precursor cells (OPCs) are recognized as the progenitors responsible for the generation of oligodendrocytes, which play a critical role in myelination. Extensive research has shed light on the mechanisms underlying OPC proliferation and differentiation into mature myelin-forming oligodendrocytes. However, recent advances in the field have revealed that OPCs have multiple functions beyond their role as progenitors, exerting control over neural circuits and brain function through distinct pathways. This review aims to provide a comprehensive understanding of OPCs by first introducing their well-established features. Subsequently, we delve into the emerging roles of OPCs in modulating brain function in both healthy and diseased states. Unraveling the cellular and molecular mechanisms by which OPCs influence brain function holds great promise for identifying novel therapeutic targets for central nervous system diseases.


Subject(s)
Oligodendrocyte Precursor Cells , Oligodendrocyte Precursor Cells/metabolism , Myelin Sheath/metabolism , Brain/metabolism , Oligodendroglia/metabolism , Central Nervous System , Cell Differentiation/physiology
3.
Pflugers Arch ; 475(11): 1283-1300, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37700120

ABSTRACT

Fluorescent dyes and genetically encoded fluorescence indicators (GEFI) are common tools for visualizing concentration changes of specific ions and messenger molecules during intra- as well as intercellular communication. Using advanced imaging technologies, fluorescence indicators are a prerequisite for the analysis of physiological molecular signaling. Automated detection and analysis of fluorescence signals require to overcome several challenges, including correct estimation of fluorescence fluctuations at basal concentrations of messenger molecules, detection, and extraction of events themselves as well as proper segmentation of neighboring events. Moreover, event detection algorithms need to be sensitive enough to accurately capture localized and low amplitude events exhibiting a limited spatial extent. Here, we present two algorithms (PBasE and CoRoDe) for accurate baseline estimation and automated detection and segmentation of fluorescence fluctuations.

4.
Glia ; 70(5): 892-912, 2022 05.
Article in English | MEDLINE | ID: mdl-35064700

ABSTRACT

The disruption of the blood-brain barrier (BBB) plays a critical role in the pathology of ischemic stroke. p75 neurotrophin receptor (p75NTR ) contributes to the disruption of the blood-retinal barrier in retinal ischemia. However, whether p75NTR influences the BBB permeability after acute cerebral ischemia remains unknown. The present study investigated the role and underlying mechanism of p75NTR on BBB integrity in an ischemic stroke mouse model, middle cerebral artery occlusion (MCAO). After 24 h of MCAO, astrocytes and endothelial cells in the infarct-affected brain area up-regulated p75NTR . Genetic p75NTR knockdown (p75NTR+/- ) or pharmacological inhibition of p75NTR using LM11A-31, a selective inhibitor of p75NTR , both attenuated brain damage and BBB leakage in MCAO mice. Astrocyte-specific conditional knockdown of p75NTR mediated with an adeno-associated virus significantly ameliorated BBB disruption and brain tissue damage, as well as the neurological functions after stroke. Further molecular biological examinations indicated that astrocytic p75NTR activated NF-κB and HIF-1α signals, which upregulated the expression of MMP-9 and vascular endothelial growth factor (VEGF), subsequently leading to tight junction degradation after ischemia. As a result, increased leukocyte infiltration and microglia activation exacerbated brain injury after stroke. Overall, our results provide novel insight into the role of astrocytic p75NTR in BBB disruption after acute cerebral ischemia. The p75NTR may therefore be a potential therapeutic target for the treatment of ischemic stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Receptors, Nerve Growth Factor/metabolism , Stroke , Animals , Astrocytes/metabolism , Blood-Brain Barrier/pathology , Brain Ischemia/metabolism , Endothelial Cells/metabolism , Infarction, Middle Cerebral Artery/pathology , Mice , Stroke/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Glia ; 67(6): 1094-1103, 2019 06.
Article in English | MEDLINE | ID: mdl-30724411

ABSTRACT

In the central nervous system, the type I transmembrane glycoprotein NG2 (nerve-glia antigen 2) is only expressed by pericytes and oligodendrocyte precursor cells (OPCs). Therefore, OPCs are also termed NG2 glia. Their fate during development has been investigated systematically in several genetically modified mouse models. Consensus exists that postnatal NG2 glia are restricted to the oligodendrocyte (OL) lineage, while, at least in the forebrain, embryonic NG2 glia could also generate astrocytes. In addition, experimental evidence for a neurogenic potential of NG2 glia in the early embryonic brain (before E16.5) has been provided. However, this observation is still controversial. Here, we took advantage of reliable transgene expression in NG2-EYFP and NG2-CreERT2 knock-in mice to study the fate of early embryonic NG2 glia. While pericytes were the main cells with robust NG2 gene activity at E12.5, only a few OPCs expressed NG2 at this early stage of embryogenesis. Subsequently, this proportion of OPCs increased from 3% (E12.5) to 11% and 25% at E14.5 and E17.5, respectively. When Cre DNA recombinase activity was induced at E12.5 and E14.5 and pups were analyzed at postnatal day 0 (P0) and P10, the vast majority of recombined cells, besides pericytes, belonged to the OL lineage cells, with few astrocytes in the ventral forebrain. In other brain regions such as brain stem, cerebellum, and olfactory bulb only OL lineage cells were detected. Therefore, we conclude that NG2 glia from early embryonic brain are restricted to a gliogenic fate and do not differentiate into neurons after birth.


Subject(s)
Antigens/biosynthesis , Brain/embryology , Brain/metabolism , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , Proteoglycans/biosynthesis , Animals , Brain Chemistry/physiology , Cell Lineage/physiology , Embryonic Development/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/chemistry , Neurons/chemistry
6.
Glia ; 65(9): 1535-1549, 2017 09.
Article in English | MEDLINE | ID: mdl-28636132

ABSTRACT

Following brain injury astrocytes change into a reactive state, proliferate and grow into the site of lesion, a process called astrogliosis, initiated and regulated by changes in cytoplasmic Ca2+ . Transient receptor potential canonical (TRPC) channels may contribute to Ca2+ influx but their presence and possible function in astrocytes is not known. By RT-PCR and RNA sequencing we identified transcripts of Trpc1, Trpc2, Trpc3, and Trpc4 in FACS-sorted glutamate aspartate transporter (GLAST)-positive cultured mouse cortical astrocytes and subcloned full-length Trpc1 and Trpc3 cDNAs from these cells. Ca2+ entry in cortical astrocytes depended on TRPC3 and was increased in the absence of Trpc1. After co-expression of Trpc1 and Trpc3 in HEK-293 cells both proteins co-immunoprecipitate and form functional heteromeric channels, with TRPC1 reducing TRPC3 activity. In vitro, lack of Trpc3 reduced astrocyte proliferation and migration whereas the TRPC3 gain-of-function moonwalker mutation and Trpc1 deficiency increased astrocyte migration. In vivo, astrogliosis and cortex edema following stab wound injury were reduced in Trpc3-/- but increased in Trpc1-/- mice. In summary, our results show a decisive contribution of TRPC3 to astrocyte Ca2+ signaling, which is even augmented in the absence of Trpc1, in particular following brain injury. Targeted therapies to reduce TRPC3 channel activity in astrocytes might therefore be beneficial in traumatic brain injury.


Subject(s)
Astrocytes/metabolism , Calcium Signaling/physiology , Cerebral Cortex/injuries , Gliosis/metabolism , TRPC Cation Channels/metabolism , Animals , Astrocytes/pathology , Brain Edema/etiology , Brain Edema/metabolism , Brain Edema/pathology , Cell Movement/physiology , Cell Proliferation/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Gliosis/etiology , Gliosis/pathology , HEK293 Cells , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Wounds, Stab/metabolism , Wounds, Stab/pathology
7.
Neurochem Res ; 42(9): 2479-2489, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28702713

ABSTRACT

An acute brain injury is commonly characterized by an extended cellular damage. The post-injury process of scar formation is largely determined by responses of various local glial cells and blood-derived immune cells. The role of astrocytes and microglia have been frequently reviewed in the traumatic sequelae. Here, we summarize the diverse contributions of oligodendrocytes (OLs) and their precursor cells (OPCs) in acute injuries. OLs at the lesion site are highly sensitive to a damaging insult, provoked by Ca2+ overload after hyperexcitation originating from increased levels of transmitters. At the lesion site, differentiating OPCs can replace injured oligodendrocytes to guarantee proper myelination that is instrumental for healthy brain function. In contrast to finally differentiated and non-dividing OLs, OPCs are the most proliferative cells of the brain and their proliferation rate even increases after injury. There exist even evidence that OPCs might also generate some type of astrocyte beside OLs. Thereby, OPCs can contribute to the generation and maintenance of the glial scar. In the future, detailed knowledge of the molecular cues that help to prevent injury-evoked glial cell death and that control differentiation and myelination of the oligodendroglial lineage will be pivotal in developing novel therapeutic approaches.


Subject(s)
Brain Injuries/pathology , Cell Lineage/physiology , Oligodendroglia/physiology , Animals , Astrocytes/pathology , Astrocytes/physiology , Brain/pathology , Brain/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Humans , Neuroglia/pathology , Neuroglia/physiology , Oligodendroglia/pathology
8.
BMC Cancer ; 16: 72, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26856327

ABSTRACT

BACKGROUND: Glioblastoma multiforme is the most aggressive brain tumor. Microglia are prominent cells within glioma tissue and play important roles in tumor biology. This work presents an animal model designed for the study of microglial cell morphology in situ during gliomagenesis. It also allows a quantitative morphometrical analysis of microglial cells during their activation by glioma cells. METHODS: The animal model associates the following cell types: 1- mCherry red fluorescent GL261 glioma cells and; 2- EGFP fluorescent microglia, present in the TgH(CX3CR1-EGFP) mouse line. First, mCherry-GL261 glioma cells were implanted in the brain cortex of TgH(CX3CR1-EGFP) mice. Epifluorescence - and confocal laser-scanning microscopy were employed for analysis of fixed tissue sections, whereas two-photon laser-scanning microscopy (2P-LSM) was used to track tumor cells and microglia in the brain of living animals. RESULTS: Implanted mCherry-GL261 cells successfully developed brain tumors. They mimic the aggressive behavior found in human disease, with a rapid increase in size and the presence of secondary tumors apart from the injection site. As tumor grows, mCherry-GL261 cells progressively lost their original shape, adopting a heterogeneous and diffuse morphology at 14-18 d. Soma size increased from 10-52 µm. At this point, we focused on the kinetics of microglial access to glioma tissues. 2P-LSM revealed an intense microgliosis in brain areas already shortly after tumor implantation, i.e. at 30 min. By confocal microscopy, we found clusters of microglial cells around the tumor mass in the first 3 days. Then cells infiltrated the tumor area, where they remained during all the time points studied, from 6-18 days. Microglia in contact with glioma cells also present changes in cell morphology, from a ramified to an amoeboid shape. Cell bodies enlarged from 366 ± 0.0 µm(2), in quiescent microglia, to 1310 ± 146.0 µm(2), and the cell processes became shortened. CONCLUSIONS: The GL261/CX3CR1 mouse model reported here is a valuable tool for imaging of microglial cells during glioma growth, either in fixed tissue sections or living animals. Remarkable advantages are the use of immunocompetent animals and the simplified imaging method without the need of immunohistochemical procedures.


Subject(s)
Cerebral Cortex/ultrastructure , Glioblastoma/ultrastructure , Glioma/ultrastructure , Animals , CX3C Chemokine Receptor 1 , Cell Line, Tumor , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , ErbB Receptors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/genetics , Glioma/pathology , Humans , Mice , Microglia/metabolism , Microglia/pathology , Microglia/ultrastructure , Microscopy, Confocal , Receptors, Chemokine/genetics
9.
Glia ; 62(6): 896-913, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24578301

ABSTRACT

NG2 (nerve/glia antigen-2) is a type I transmembrane glycoprotein and also known as chondroitin sulfate proteoglycan 4. In the parenchyma of the central nervous system, NG2-expressing (NG2(+) ) cells have been identified as a novel type of glia with a strong potential to generate oligodendrocytes (OLs) in the developing white matter. However, the differentiation potential of NG2 glia remained controversial, largely attributable to shortcomings of transgenic mouse models used for fate mapping. To minimize these restrictions and to more faithfully mimic the endogenous NG2 expression in vivo, we generated a mouse line in which the open reading frame of the tamoxifen-inducible form of the Cre DNA recombinase (CreERT2) was inserted into the NG2 locus by homologous recombination. Results from this novel mouse line demonstrate that at different developmental stages of the brain, NG2(+) cells either stayed as NG2 glia or differentiated into OLs during the whole life span. Interestingly, when Cre activity was induced at embryonic stages, a significant number of reporter(+) astrocytes could be detected in the gray matter after birth. However, in other brain regions, such as olfactory bulb, brain stem, and cerebellum, all of the NG2 glia was restricted to the OL lineage. In addition, tamoxifen-sensitive and NG2 gene locus-dependent gene recombination could be detected in a small, but persistent population of cortical NeuN(+) neurons starting from the second postnatal week.


Subject(s)
Antigens/biosynthesis , Antigens/genetics , Cell Differentiation/physiology , Integrases/biosynthesis , Integrases/genetics , Neuroglia/physiology , Proteoglycans/biosynthesis , Proteoglycans/genetics , Animals , Female , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendroglia/physiology , Pregnancy
10.
Front Immunol ; 15: 1425706, 2024.
Article in English | MEDLINE | ID: mdl-39044821

ABSTRACT

Oligodendrocyte precursor cells (OPCs) have long been regarded as progenitors of oligodendrocytes, yet recent advances have illuminated their multifaceted nature including their emerging immune functions. This review seeks to shed light on the immune functions exhibited by OPCs, spanning from phagocytosis to immune modulation and direct engagement with immune cells across various pathological scenarios. Comprehensive understanding of the immune functions of OPCs alongside their other roles will pave the way for targeted therapies in neurological disorders.


Subject(s)
Oligodendrocyte Precursor Cells , Humans , Oligodendrocyte Precursor Cells/immunology , Animals , Phagocytosis/immunology , Oligodendroglia/immunology , Cell Differentiation/immunology , Immunomodulation
11.
Nat Commun ; 15(1): 6340, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068155

ABSTRACT

Molecular pathways mediating systemic inflammation entering the brain parenchyma to induce sepsis-associated encephalopathy (SAE) remain elusive. Here, we report that in mice during the first 6 hours of peripheral lipopolysaccharide (LPS)-evoked systemic inflammation (6 hpi), the plasma level of adenosine quickly increased and enhanced the tone of central extracellular adenosine which then provoked neuroinflammation by triggering early astrocyte reactivity. Specific ablation of astrocytic Gi protein-coupled A1 adenosine receptors (A1ARs) prevented this early reactivity and reduced the levels of inflammatory factors (e.g., CCL2, CCL5, and CXCL1) in astrocytes, thereby alleviating microglial reaction, ameliorating blood-brain barrier disruption, peripheral immune cell infiltration, neuronal dysfunction, and depression-like behaviour in the mice. Chemogenetic stimulation of Gi signaling in A1AR-deficent astrocytes at 2 and 4 hpi of LPS injection could restore neuroinflammation and depression-like behaviour, highlighting astrocytes rather than microglia as early drivers of neuroinflammation. Our results identify early astrocyte reactivity towards peripheral and central levels of adenosine as an important pathway driving SAE and highlight the potential of targeting A1ARs for therapeutic intervention.


Subject(s)
Adenosine , Astrocytes , Lipopolysaccharides , Mice, Inbred C57BL , Microglia , Receptor, Adenosine A1 , Sepsis-Associated Encephalopathy , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/immunology , Adenosine/metabolism , Mice , Sepsis-Associated Encephalopathy/metabolism , Receptor, Adenosine A1/metabolism , Male , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Disease Models, Animal , Sepsis/immunology , Sepsis/complications , Neuroinflammatory Diseases/immunology , Brain/metabolism , Brain/pathology , Brain/immunology , Brain/drug effects , Mice, Knockout , Inflammation , Signal Transduction/drug effects
12.
Nat Commun ; 15(1): 2866, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570482

ABSTRACT

Traumatic brain injury leads to a highly orchestrated immune- and glial cell response partially responsible for long-lasting disability and the development of secondary neurodegenerative diseases. A holistic understanding of the mechanisms controlling the responses of specific cell types and their crosstalk is required to develop an efficient strategy for better regeneration. Here, we combine spatial and single-cell transcriptomics to chart the transcriptomic signature of the injured male murine cerebral cortex, and identify specific states of different glial cells contributing to this signature. Interestingly, distinct glial cells share a large fraction of injury-regulated genes, including inflammatory programs downstream of the innate immune-associated pathways Cxcr3 and Tlr1/2. Systemic manipulation of these pathways decreases the reactivity state of glial cells associated with poor regeneration. The functional relevance of the discovered shared signature of glial cells highlights the importance of our resource enabling comprehensive analysis of early events after brain injury.


Subject(s)
Brain Injuries , Wounds, Stab , Animals , Mice , Male , Glial Fibrillary Acidic Protein/metabolism , Neuroglia/metabolism , Brain Injuries/metabolism , Cerebral Cortex/metabolism , Wounds, Stab/complications , Wounds, Stab/metabolism
13.
Dev Cell ; 58(13): 1153-1169.e5, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37220747

ABSTRACT

Acute brain injuries evoke various response cascades directing the formation of the glial scar. Here, we report that acute lesions associated with hemorrhagic injuries trigger a re-programming of oligodendrocytes. Single-cell RNA sequencing highlighted a subpopulation of oligodendrocytes activating astroglial genes after acute brain injuries. By using PLP-DsRed1/GFAP-EGFP and PLP-EGFPmem/GFAP-mRFP1 transgenic mice, we visualized this population of oligodendrocytes that we termed AO cells based on their concomitant activity of astro- and oligodendroglial genes. By fate mapping using PLP- and GFAP-split Cre complementation and repeated chronic in vivo imaging with two-photon laser-scanning microscopy, we observed the conversion of oligodendrocytes into astrocytes via the AO cell stage. Such conversion was promoted by local injection of IL-6 and was diminished by IL-6 receptor-neutralizing antibody as well as by inhibiting microglial activation with minocycline. In summary, our findings highlight the plastic potential of oligodendrocytes in acute brain trauma due to microglia-derived IL-6.


Subject(s)
Astrocytes , Brain Injuries , Mice , Animals , Interleukin-6 , Glial Fibrillary Acidic Protein/genetics , Oligodendroglia , Mice, Transgenic
14.
Nat Commun ; 13(1): 1394, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35296664

ABSTRACT

Cortical neural circuits are complex but very precise networks of balanced excitation and inhibition. Yet, the molecular and cellular mechanisms that form the balance are just beginning to emerge. Here, using conditional γ-aminobutyric acid receptor B1- deficient mice we identify a γ-aminobutyric acid/tumor necrosis factor superfamily member 12-mediated bidirectional communication pathway between parvalbumin-positive fast spiking interneurons and oligodendrocyte precursor cells that determines the density and function of interneurons in the developing medial prefrontal cortex. Interruption of the GABAergic signaling to oligodendrocyte precursor cells results in reduced myelination and hypoactivity of interneurons, strong changes of cortical network activities and impaired social cognitive behavior. In conclusion, glial transmitter receptors are pivotal elements in finetuning distinct brain functions.


Subject(s)
Oligodendrocyte Precursor Cells , Animals , Cognition , Communication , Interneurons/physiology , Mice , Oligodendrocyte Precursor Cells/metabolism , Parvalbumins/metabolism
15.
Neurosci Bull ; 37(7): 1039-1050, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33928492

ABSTRACT

GABA is the main inhibitory neurotransmitter in the CNS acting at two distinct types of receptor: ligand-gated ionotropic GABAA receptors and G protein-coupled metabotropic GABAB receptors, thus mediating fast and slow inhibition of excitability at central synapses. GABAergic signal transmission has been intensively studied in neurons in contrast to oligodendrocytes and their precursors (OPCs), although the latter express both types of GABA receptor. Recent studies focusing on interneuron myelination and interneuron-OPC synapses have shed light on the importance of GABA signaling in the oligodendrocyte lineage. In this review, we start with a short summary on GABA itself and neuronal GABAergic signaling. Then, we elaborate on the physiological role of GABA receptors within the oligodendrocyte lineage and conclude with a description of these receptors as putative targets in treatments of CNS diseases.


Subject(s)
Neurons , Oligodendroglia , Receptors, GABA , Receptors, GABA-A , Synapses
16.
Brain Behav Immun ; 24(4): 585-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20083190

ABSTRACT

The central nervous system (CNS) does not regenerate partly due to the slow clearance of debris from the degenerated myelin sheath by Wallerian degeneration. The mechanism underlying the inefficiency in myelin clearance is not clear. Here we showed that endogenous proBDNF may inhibit the infiltration of ED1+ inflammatory cells after spinal cord injury. After injury, proBDNF and its receptors sortilin and p75NTR are expressed in the spinal cord as determined by Western blots and immunocytochemistry. ProBDNF and mature BDNF were released from macrophages in vitro. Macrophages in vivo (ED1+) and isolated in vitro (CD11b+) express moderate levels of proBDNF, sortilin and p75NTR. ProBDNF suppressed the migration of isolated macrophages in vitro and the antibody to proBDNF enhanced the migration. Suppression of proBDNF in vivo by administering the antiserum to the prodomain of BDNF after spinal cord injury (SCI) increased the infiltration of macrophages and increased number of neurons in the injured cord. BBB tests showed that the treatment of the antibody to proBDNF improved the functional recovery after spinal cord injury. Our data suggest that proBDNF is a suppressing factor for macrophage migration and infiltration and may play a detrimental role after SCI.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cell Migration Inhibition/immunology , Macrophages/immunology , Receptors, Nerve Growth Factor/metabolism , Spinal Cord Injuries/immunology , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/immunology , CD11b Antigen , Cells, Cultured , Disease Models, Animal , Ectodysplasins , Female , Immunohistochemistry , Macrophages/metabolism , Nerve Tissue Proteins , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor , Recovery of Function , Spinal Cord/cytology , Spinal Cord Injuries/metabolism
17.
Acta Neuropathol Commun ; 8(1): 146, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32843103

ABSTRACT

NG2 is a type I transmembrane glycoprotein known as chondroitin sulfate proteoglycan 4 (CSPG4). In the healthy central nervous system, NG2 is exclusively expressed by oligodendrocyte progenitor cells and by vasculature pericytes. A large body of immunohistochemical studies showed that under pathological conditions such as acute brain injuries and experimental autoimmune encephalomyelitis (EAE), a number of activated microglia were NG2 immuno-positive, suggesting NG2 expression in these cells. Alternative explanations for the microglial NG2 labeling consider the biochemical properties of NG2 or the phagocytic activity of activated microglia. Reportedly, the transmembrane NG2 proteoglycan can be cleaved by a variety of proteases to deposit the NG2 ectodomain into the extracellular matrix. The ectodomain, however, could also stick to the microglial surface. Since microglia are phagocytic cells engulfing debris of dying cells, it is difficult to identify a genuine expression of NG2. Recent studies showing (1) pericytes giving rise to microglial after stroke, and (2) immune cells of NG2-EYFP knock-in mice lacking NG2 expression in an EAE model generated doubts for the de novo expression of NG2 in microglia after acute brain injuries. In the current study, we took advantage of three knock-in mouse lines (NG2-CreERT2, CX3CR1-EGFP and NG2-EYFP) to study NG2 expression indicated by transgenic fluorescent proteins in microglia after tMCAO (transient middle cerebral artery occlusion) or cortical stab wound injury (SWI). We provide strong evidence that NG2-expressing cells, including OPCs and pericytes, did not differentiate into microglia after acute brain injuries, whereas activated microglia did express NG2 in a disease-dependent manner. A subset of microglia continuously activated the NG2 gene at least within the first week after tMCAO, whereas within 3 days after SWI a limited number of microglia at the lesion site transiently expressed NG2. Immunohistochemical studies demonstrated that these microglia with NG2 gene activity also synthesized the NG2 protein, suggesting activated microglia as an additional source of the NG2 proteoglycan after acute brain injuries.


Subject(s)
Antigens/metabolism , Brain Injuries/metabolism , Microglia/metabolism , Proteoglycans/metabolism , Animals , Gene Knock-In Techniques , Mice , Mice, Transgenic
18.
Acta Physiol (Oxf) ; : e14225, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39221537
20.
Neural Regen Res ; 18(12): 2649-2650, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37449605
SELECTION OF CITATIONS
SEARCH DETAIL