Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Brain Commun ; 4(2): fcac005, 2022.
Article in English | MEDLINE | ID: mdl-35282162

ABSTRACT

Demyelination of the central nervous system is a prominent pathological hallmark of multiple sclerosis and affects both white and grey matter. However, demyelinated white and grey matter exhibit clear pathological differences, most notably the presence or absence of inflammation and activated glial cells in white and grey matter, respectively. In order to gain more insight into the differential pathology of demyelinated white and grey matter areas, we micro-dissected neighbouring white and grey matter demyelinated areas as well as normal-appearing matter from leucocortical lesions of human post-mortem material and used these samples for RNA sequencing. Our data show that even neighbouring demyelinated white and grey matter of the same leucocortical have a distinct gene expression profile and cellular composition. We propose that, based on their distinct expression profile, pathological processes in neighbouring white and grey matter are likely different which could have implications for the efficacy of treating grey matter lesions with current anti-inflammatory-based multiple sclerosis drugs.

2.
Brain Behav Immun Health ; 15: 100265, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34589771

ABSTRACT

Microglia are tissue-resident macrophages of the central nervous system (CNS), and important for CNS development and homeostasis. In the adult CNS, microglia monitor environmental changes and react to tissue damage, cellular debris, and pathogens. Here, we present a gene expression profile of purified microglia isolated from the rhesus macaque, a non-human primate, that consists of 666 transcripts. The macaque microglia transcriptome was intersected with the transcriptional programs of microglia from mouse, zebrafish, and human CNS tissues, to determine (dis)similarities. This revealed an extensive overlap of 342 genes between the transcriptional profile of macaque and human microglia, and showed that the gene expression profile of zebrafish is most distant when compared to other species. Furthermore, an evolutionair core based on the overlapping gene expression signature from all four species was identified. This study presents a macaque microglia transcriptomics profile, and identifies a gene expression program in microglia that is preserved across species, underscoring their CNS-tailored tissue macrophage functions as innate immune cells with CNS-surveilling properties.

3.
Annu Rev Physiol ; 79: 619-643, 2017 02 10.
Article in English | MEDLINE | ID: mdl-27959620

ABSTRACT

As the immune-competent cells of the brain, microglia play an increasingly important role in maintaining normal brain function. They invade the brain early in development, transform into a highly ramified phenotype, and constantly screen their environment. Microglia are activated by any type of pathologic event or change in brain homeostasis. This activation process is highly diverse and depends on the context and type of the stressor or pathology. Microglia can strongly influence the pathologic outcome or response to a stressor due to the release of a plethora of substances, including cytokines, chemokines, and growth factors. They are the professional phagocytes of the brain and help orchestrate the immunological response by interacting with infiltrating immune cells. We describe here the diversity of microglia phenotypes and their responses in health, aging, and disease. We also review the current literature about the impact of lifestyle on microglia responses and discuss treatment options that modulate microglial phenotypes.


Subject(s)
Brain/immunology , Microglia/immunology , Microglia/physiology , Aging/immunology , Aging/physiology , Animals , Brain/physiology , Humans , Phagocytes/immunology , Phagocytes/physiology
4.
Glia ; 65(1): 50-61, 2017 01.
Article in English | MEDLINE | ID: mdl-27615381

ABSTRACT

Huntington's disease (HD) is an autosomal dominant inherited neurodegenerative disorder that is caused by a CAG expansion in the Huntingtin (HTT) gene, leading to HTT inclusion formation in the brain. The mutant huntingtin protein (mHTT) is ubiquitously expressed and therefore nuclear inclusions could be present in all brain cells. The effects of nuclear inclusion formation have been mainly studied in neurons, while the effect on glia has been comparatively disregarded. Astrocytes, microglia, and oligodendrocytes are glial cells that are essential for normal brain function and are implicated in several neurological diseases. Here we examined the number of nuclear mHTT inclusions in both neurons and various types of glia in the two brain areas that are the most affected in HD, frontal cortex, and striatum. We compared nuclear mHTT inclusion body formation in three HD mouse models that express either full-length HTT or an N-terminal exon1 fragment of mHTT, and we observed nuclear inclusions in neurons, astrocytes, oligodendrocytes, and microglia. When studying the frequency of cells with nuclear inclusions in mice, we found that half of the population of neurons contained nuclear inclusions at the disease end stage, whereas the proportion of GFAP-positive astrocytes and oligodendrocytes having a nuclear inclusion was much lower, while microglia hardly showed any nuclear inclusions. Nuclear inclusions were also present in neurons and all studied glial cell types in human patient material. This is the first report to compare nuclear mHTT inclusions in glia and neurons in different HD mouse models and HD patient brains. GLIA 2016;65:50-61.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/genetics , Neuroglia/metabolism , Neurons/metabolism , Animals , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Disease Models, Animal , Female , Huntington Disease/metabolism , Male , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism
5.
Exp Brain Res ; 235(3): 731-742, 2017 03.
Article in English | MEDLINE | ID: mdl-27866262

ABSTRACT

The formation of oligomers and aggregates of overexpressed or mutant α-synuclein play a role in the degeneration of dopaminergic neurons in Parkinson's disease by causing dysfunction of mitochondria, reflected in their disturbed mobility and production of ROS. The mode of action and mechanisms underlying this mitochondrial impairment is still unclear. We have induced stable expression of wild-type, A30P or A53T α-synuclein in neuronally differentiated SH-SY5Y neuroblastoma cells and studied anterograde and retrograde mitochondrial trafficking in this cell model for Parkinson's disease. In contrast to wild-type and A30P, A53T α-synuclein significantly inhibited mitochondrial trafficking, at first retrogradely and in a later stage anterogradely. Accordingly, A53T α-synuclein also caused the highest increase in ROS production in the dysmobilized mitochondria in comparison to wild-type or A30P α-synuclein. Treatment with NAP, the eight amino acid peptide identified as the active component of activity-dependent neuroprotective protein (ADNP), completely annihilated the adverse effects of A53T on mitochondrial dynamics. Our results reveal that A53T α-synuclein (oligomers or aggregates) leads to the inhibition of mitochondrial trafficking, which can be rescued by NAP, suggesting the involvement of microtubule disruption in the pathophysiology of Parkinson's disease.


Subject(s)
Mitochondria/drug effects , Mitochondria/genetics , Oligopeptides/pharmacology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Alanine/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Kymography , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Threonine/genetics , Transfection , alpha-Synuclein/chemistry
6.
Oncotarget ; 6(31): 31007-17, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26307681

ABSTRACT

Differentiation has been proposed as a therapeutic strategy for glioblastoma (GBM) in part due to observations of stem-like cells in GBM that have been shown to undergo terminal differentiation in response to growth factor withdrawal and BMP activation. However, the effects of long term exposure to serum culture conditions on glioma sphere cultures/glioma stem-like cells (GSCs) have not been examined. Here we show that GSCs retained both neurosphere formation and tumor initiation abilities after short or long term serum exposure. Under these conditions, GSCs expressed both neural lineage and stem cell markers, highlighting the aberrant pseudo-differentiation state. GSCs maintained under adherent serum cultured conditions continued to proliferate and initiate tumor formation with efficiencies similar to GSCs maintained under proliferating (neurosphere) conditions. Proneural (PN) GSCs under serum exposure showed an induction of mesenchymal (MES) gene expression signatures. Our data indicate that exposure to serum containing media result in aberrant differentiation (e.g. toward MES lineage) and activation of alternative oncogenic pathways in GSCs.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Cell Transformation, Neoplastic/pathology , Glioma/pathology , Mesoderm/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Mesoderm/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Brain Behav Immun ; 48: 205-21, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25843371

ABSTRACT

Microglia, the innate immune cells of the central nervous system (CNS), react to endotoxins like bacterial lipopolysaccharides (LPS) with a pronounced inflammatory response. To avoid excess damage to the CNS, the microglia inflammatory response needs to be tightly regulated. Here we report that a single LPS challenge results in a prolonged blunted pro-inflammatory response to a subsequent LPS stimulation, both in primary microglia cultures (100 ng/ml) and in vivo after intraperitoneal (0.25 and 1mg/kg) or intracerebroventricular (5 µg) LPS administration. Chromatin immunoprecipitation (ChIP) experiments with primary microglia and microglia acutely isolated from mice showed that LPS preconditioning was accompanied by a reduction in active histone modifications AcH3 and H3K4me3 in the promoters of the IL-1ß and TNF-α genes. Furthermore, LPS preconditioning resulted in an increase in the amount of repressive histone modification H3K9me2 in the IL-1ß promoter. ChIP and knock-down experiments showed that NF-κB subunit RelB was bound to the IL-1ß promoter in preconditioned microglia and that RelB is required for the attenuated LPS response. In addition to a suppressed pro-inflammatory response, preconditioned primary microglia displayed enhanced phagocytic activity, increased outward potassium currents and nitric oxide production in response to a second LPS challenge. In vivo, a single i.p. LPS injection resulted in reduced performance in a spatial learning task 4 weeks later, indicating that a single inflammatory episode affected memory formation in these mice. Summarizing, we show that LPS-preconditioned microglia acquire an epigenetically regulated, immune-suppressed phenotype, possibly to prevent excessive damage to the central nervous system in case of recurrent (peripheral) inflammation.


Subject(s)
Epigenesis, Genetic , Gene Silencing , Lipopolysaccharides/pharmacology , Microglia/metabolism , Transcription Factor RelB/metabolism , Animals , Histones/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Microglia/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Glia ; 62(10): 1724-35, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24953459

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS) characterized by loss of myelin accompanied by infiltration of T-lymphocytes and monocytes. Although it has been shown that these infiltrates are important for the progression of MS, the role of microglia, the resident macrophages of the CNS, remains ambiguous. Therefore, we have compared the phenotypes of microglia and macrophages in a mouse model for MS, experimental autoimmune encephalomyelitis (EAE). In order to properly discriminate between these two cell types, microglia were defined as CD11b(pos) CD45(int) Ly-6C(neg) , and infiltrated macrophages as CD11b(pos) CD45(high) Ly-6C(pos) . During clinical EAE, microglia displayed a weakly immune-activated phenotype, based on the expression of MHCII, co-stimulatory molecules (CD80, CD86, and CD40) and proinflammatory genes [interleukin-1ß (IL-1ß) and tumour necrosis factor- α (TNF-α)]. In contrast, CD11b(pos) CD45(high) Ly-6C(pos) infiltrated macrophages were strongly activated and could be divided into two populations Ly-6C(int) and Ly-6C(high) , respectively. Ly-6C(high) macrophages contained less myelin than Ly-6C(int) macrophages and expression levels of the proinflammatory cytokines IL-1ß and TNF-α were higher in Ly-6C(int) macrophages. Together, our data show that during clinical EAE, microglia are only weakly activated whereas infiltrated macrophages are highly immune reactive.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophages/immunology , Microglia/immunology , Animals , Antigens, Ly/metabolism , CD11b Antigen/metabolism , Caspase 6/metabolism , Chimera , Cytokines/metabolism , Disease Models, Animal , Female , Interleukin-1beta/metabolism , Leukocyte Common Antigens/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis , Spinal Cord/immunology
9.
J Neuroimmune Pharmacol ; 8(4): 807-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23881706

ABSTRACT

Microglia are the prime innate immune cells of the central nervous system. They can transit from a (so-called) resting state under homeostatic conditions towards a pro-inflammatory activation state upon homeostatic disturbances. Under neurodegenerative conditions, microglia have been largely perceived as neurotoxic cells. It is now becoming clear that resting microglia are not inactive but that they serve house-keeping functions. Moreover, microglia activity is not limited to proinflammatory responses, but covers a spectrum of reactive profiles. Depending on the actual situation, activated microglia display specific effector functions supporting inflammation, tissue remodeling, synaptic plasticity and neurogenesis. Many of these functions not only relate to the current state of the local neural environment but also depend on previous experience. In this review, we address microglia functions with respect to determining factors, phenotypic presentations, adaptation to environmental signals and aging. Finally, we point out primary mechanisms of microglia activation, which may comprise therapeutic targets to control neuro-inflammatory and neurodegenerative activity.


Subject(s)
Adaptation, Physiological , Microglia/physiology , Phenotype , Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Aging/genetics , Aging/immunology , Aging/pathology , Animals , Humans , Microglia/immunology , Microglia/pathology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology
10.
Br J Pharmacol ; 168(6): 1375-87, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23121557

ABSTRACT

BACKGROUND AND PURPOSE: Induction of cellular migration is the primary effect of chemokine receptor activation. However, several chemokine receptor-like proteins bind chemokines without subsequent induction of intracellular signalling and chemotaxis. It has been suggested that they act as chemokine scavengers, which may control local chemokine levels and contribute to the function of chemokines during inflammation. This has been verified for the chemokine-like receptor proteins D6 and DARC as well as CCX-CKR. Here, we provide evidence for an additional biological function of human (h)CCX-CKR. EXPERIMENTAL APPROACH: We used transfection strategies in HEK293 and human T cells. KEY RESULTS: Co-expression of hCCX-CKR completely inhibits hCXCR3-induced chemotaxis. We found that hCCX-CKR forms complexes with hCXCR3, suggesting a relationship between CCX-CKR heteromerization and inhibition of chemotaxis. Moreover, negative binding cooperativity induced by ligands both for hCXCR3 and hCCX-CKR was observed in cells expressing both receptors. This negative cooperativity may also explain the hCCX-CKR-induced inhibition of chemotaxis. CONCLUSIONS AND IMPLICATIONS: These findings suggest that hCCX-CKR prevents hCXCR3-induced chemotaxis by heteromerization thus representing a novel mechanism of regulation of immune cell migration.


Subject(s)
Chemotaxis, Leukocyte , Down-Regulation , Receptors, CCR/metabolism , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology , Cells, Cultured , Chemokines/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation , HEK293 Cells , Humans , Immunohistochemistry , Kinetics , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Protein Multimerization , Protein Transport , RNA, Messenger , Receptors, CCR/genetics , Receptors, CXCR3/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
11.
Br J Pharmacol ; 168(7): 1662-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23170857

ABSTRACT

BACKGROUND AND PURPOSE: The C-X-C chemokine receptors 3 (CXCR3) and C-X-C chemokine receptors 4 (CXCR4) are involved in various autoimmune diseases and cancers. Small antagonists have previously been shown to cross-inhibit chemokine binding to CXCR4, CC chemokine receptors 2 (CCR2) and 5 (CCR5) heteromers. We investigated whether CXCR3 and CXCR4 can form heteromeric complexes and the binding characteristics of chemokines and small ligand compounds to these chemokine receptor heteromers. EXPERIMENTAL APPROACH: CXCR3-CXCR4 heteromers were identified in HEK293T cells using co-immunoprecipitation, time-resolved fluorescence resonance energy transfer, saturation BRET and the GPCR-heteromer identification technology (HIT) approach. Equilibrium competition binding and dissociation experiments were performed to detect negative binding cooperativity. KEY RESULTS: We provide evidence that chemokine receptors CXCR3 and CXCR4 form heteromeric complexes in HEK293T cells. Chemokine binding was mutually exclusive on membranes co-expressing CXCR3 and CXCR4 as revealed by equilibrium competition binding and dissociation experiments. The small CXCR3 agonist VUF10661 impaired binding of CXCL12 to CXCR4, whereas small antagonists were unable to cross-inhibit chemokine binding to the other chemokine receptor. In contrast, negative binding cooperativity between CXCR3 and CXCR4 chemokines was not observed in intact cells. However, using the GPCR-HIT approach, we have evidence for specific ß-arrestin2 recruitment to CXCR3-CXCR4 heteromers in response to agonist stimulation. CONCLUSIONS AND IMPLICATIONS: This study indicates that heteromeric CXCR3-CXCR4 complexes may act as functional units in living cells, which potentially open up novel therapeutic opportunities.


Subject(s)
Receptors, CXCR3/metabolism , Receptors, CXCR4/metabolism , Arrestins/metabolism , Cell Membrane/metabolism , Chemokine CXCL10/metabolism , Chemokine CXCL12/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Immunoprecipitation , Ligands , Protein Binding , Protein Multimerization , Radioligand Assay , Receptors, CXCR3/agonists , Receptors, CXCR4/agonists , Signal Transduction , beta-Arrestins
12.
Neuropathol Appl Neurobiol ; 38(1): 39-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21696420

ABSTRACT

AIMS: HSPB8 is a small heat shock protein that forms a complex with the co-chaperone BAG3. Overexpression of the HSPB8-BAG3 complex in cells stimulates autophagy and facilitates the clearance of mutated aggregation-prone proteins, whose accumulation is a hallmark of many neurodegenerative disorders. HSPB8-BAG3 could thus play a protective role in protein aggregation diseases and might be specifically upregulated in response to aggregate-prone protein-mediated toxicity. Here we analysed HSPB8-BAG3 expression levels in post-mortem human brain tissue from patients suffering of the following protein conformation disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease and spinocerebellar ataxia type 3 (SCA3). METHODS: Western blotting and immunohistochemistry techniques were used to analyse HSPB8 and BAG3 expression levels in fibroblasts from SCA3 patients and post-mortem brain tissues, respectively. RESULTS: In all diseases investigated, we observed a strong upregulation of HSPB8 and a moderate upregulation of BAG3 specifically in astrocytes in the cerebral areas affected by neuronal damage and degeneration. Intriguingly, no significant change in the HSPB8-BAG3 expression levels was observed within neurones, irrespective of their localization or of the presence of proteinaceous aggregates. CONCLUSIONS: We propose that the upregulation of HSPB8 and BAG3 may enhance the ability of astrocytes to clear aggregated proteins released from neurones and cellular debris, maintain the local tissue homeostasis and/or participate in the cytoskeletal remodelling that astrocytes undergo during astrogliosis.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Astrocytes/metabolism , Heat-Shock Proteins/biosynthesis , Neurodegenerative Diseases/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Apoptosis Regulatory Proteins , Blotting, Western , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Molecular Chaperones , Up-Regulation
13.
CNS Neurol Disord Drug Targets ; 10(1): 108-18, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21143141

ABSTRACT

Microglia, the tissue macrophages of the brain, have under healthy conditions a resting phenotype that is characterized by a ramified morphology. With their fine processes microglia are continuously scanning their environment. Upon any homeostatic disturbance microglia rapidly change their phenotype and contribute to processes including inflammation, tissue remodeling, and neurogenesis. In this review, we will address functional phenotypes of microglia in diverse brain regions and phenotypes associated with neuroinflammation, neurogenesis, brain tumor homeostasis, and aging.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Inflammation/pathology , Microglia/physiology , Neurogenesis , Aging/pathology , Brain Neoplasms/physiopathology , Cells, Cultured , Humans , Phenotype
14.
Neuropathol Appl Neurobiol ; 35(6): 579-91, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19627512

ABSTRACT

AIMS: It has been shown that neural stem cells (NSCs) migrate towards areas of brain injury or brain tumours and that NSCs have the capacity to track infiltrating tumour cells. The possible mechanism behind the migratory behaviour of NSCs is not yet completely understood. As chemokines are involved in the migration of immune cells in the injured brain, they may also be involved in chemoattraction of NSCs towards a brain tumour. METHODS: The expression profile of various chemokine receptors in NSCs, harvested from the subventricular zone of adult mice, was investigated by reverse transcriptase- polymerase chain reaction analysis. Furthermore, the functionality of the chemokine receptors was assessed in in vitro chemotaxis assays and calcium signalling experiments. To test the in vivo migration of NSCs, a syngeneic mouse model was developed, whereby a B16F10 melanoma cell line was grafted into one hemisphere and later NSCs were grafted in the contralateral hemisphere. Furthermore, the expression of chemokines in this melanoma cell line was investigated. RESULTS AND CONCLUSIONS: Adult mouse NSCs functionally express various chemokine receptors of which CXC chemokine receptor (CXCR)4 shows the highest mRNA levels and most pronounced functional responses in vitro. CXC chemokine ligand (CXCL)12, the ligand for CXCR4, is expressed by the melanoma cell line. In this mouse model for metastatic brain tumours, it is shown that NSCs express CXCR4 at their cell membranes while they migrate towards the tumour, which produces CXCL12. It is therefore suggested that the CXCR4/CXCL12 pathway plays a role in the mechanism underlying tumour-mediated attraction of NSCs.


Subject(s)
Adult Stem Cells/physiology , Brain Neoplasms/physiopathology , Cell Movement/physiology , Chemokine CXCL12/metabolism , Neurons/cytology , Receptors, CXCR4/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Chemotaxis/physiology , Melanoma/physiopathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/physiopathology , Neurons/physiology , RNA, Messenger/metabolism , Receptors, CXCR/metabolism , Signal Transduction , Stem Cell Niche/physiopathology
15.
Neuropathol Appl Neurobiol ; 35(1): 89-102, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187061

ABSTRACT

AIMS: Previous studies on the therapeutic time window for intravascular administration of bone marrow stem cells (BMSCs) after stroke have shown that early intervention (from 3 h after onset) in the middle cerebral artery occlusion (MCAO) rat model is the most effective approach to reduce ischaemic lesion size. We have confirmed these observations but noticed that 2 weeks after transplantation, almost none of the grafted BMSCs could be detected in or around the lesion. The present experiments aimed to assess the fate and kinetics of intravascularly injected BMSCs shortly after administration in correlation to the development of the ischaemic lesion after MCAO. METHODS: We administered a syngeneic suspension of complete (haematopoietic and mesenchymal) BMSCs via the carotid artery to rats at 2 h after MCAO onset. We examined the distribution and tissue location of BMSCs within the first 24 h after arterial administration by perfusion-fixating rats and performing immunohistochemical analysis at different time points. RESULTS: The vast majority (>95%) of BMSCs appeared to become trapped in the spleen shortly after injection. Six hours after implantation, together with the appearance of activated microglia, the first BMSCs could be detected in and around the lesion; their number gradually increased during the first 12 h after implantation but started to decrease at 24 h. The implanted BMSCs were surrounded by activated and phagocytotic microglia. CONCLUSION: Our results show that ischaemic lesion size reduction can already be achieved by the early transient presence at the lesion site of intravascularly implanted BMSCs, possibly mediated via activated microglia.


Subject(s)
Brain Ischemia/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Animals , Apoptosis , Brain/physiopathology , Carotid Arteries , Immunohistochemistry , Infarction, Middle Cerebral Artery/therapy , Injections, Intra-Arterial , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Microglia/physiology , Phagocytosis , Rats , Rats, Wistar , Spleen/physiology , Stroke/therapy
16.
J Neuroimmunol ; 198(1-2): 69-74, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18538419

ABSTRACT

Our understanding of microglia biology has significantly changed in the last couple of years. Instead of being predominantly detrimental cells showing a stereotypic activation pattern, microglia today are considered highly adaptive elements with many distinct phenotypes. Microglia activity is aimed to protect and to restore and only in case of uncontrolled or impaired microglia function these cells may have detrimental effects. The control of microglia activity is thus an important issue to understand. The family of chemokines are versatile signals specialized to control cell-cell interactions. Neurons express chemokines in a temporarily and spatially regulated manner and microglia respond to these messengers via the appropriate receptors. Due to these features are chemokines ideal messengers for the communication between neurons and microglia.


Subject(s)
Chemokines/physiology , Microglia/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , Cell Communication/physiology
17.
Neuropsychopharmacology ; 33(9): 2237-50, 2008 Aug.
Article in English | MEDLINE | ID: mdl-17987062

ABSTRACT

The immunological response in the brain is crucial to overcome neuropathological events. Some inflammatory mediators, such as the immunoregulatory cytokine interleukin-6 (IL-6) affect neuromodulation and may also play protective roles against various noxious conditions. However, the fundamental mechanisms underlying the long-term effects of IL-6 in the brain remain unclear. We now report that IL-6 increases the expression and function of the neuronal adenosine A1 receptor, with relevant consequences to synaptic transmission and neuroprotection. IL-6-induced amplification of A1 receptor function enhances the responses to readily released adenosine during hypoxia, enables neuronal rescue from glutamate-induced death, and protects animals from chemically induced convulsing seizures. Taken together, these results suggest that IL-6 minimizes the consequences of excitotoxic episodes on brain function through the enhancement of endogenous adenosinergic signaling.


Subject(s)
Interleukin-6/pharmacology , Neurons/drug effects , Receptor, Adenosine A1/metabolism , Synaptic Transmission/drug effects , Up-Regulation/drug effects , Analysis of Variance , Animals , Autoradiography/methods , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/radiation effects , Hippocampus/drug effects , Hippocampus/physiology , Interleukin-6/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pentylenetetrazole/pharmacology , Radioligand Assay/methods , Receptor, Adenosine A1/genetics , Seizures/chemically induced , Seizures/drug therapy , Seizures/genetics , Time Factors
18.
Mol Neurobiol ; 36(2): 137-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17952658

ABSTRACT

Whereas chemokines are well known for their ability to induce cell migration, only recently it became evident that chemokines also control a variety of other cell functions and are versatile messengers in the interaction between a diversity of cell types. In the central nervous system (CNS), chemokines are generally found under both physiological and pathological conditions. Whereas many reports describe chemokine expression in astrocytes and microglia and their role in the migration of leukocytes into the CNS, only few studies describe chemokine expression in neurons. Nevertheless, the expression of neuronal chemokines and the corresponding chemokine receptors in CNS cells under physiological and pathological conditions indicates that neuronal chemokines contribute to CNS cell interaction. In this study, we review recent studies describing neuronal chemokine expression and discuss potential roles of neuronal chemokines in neuron-astrocyte, neuron-microglia, and neuron-neuron interaction.


Subject(s)
Central Nervous System , Chemokines/metabolism , Animals , Astrocytes/metabolism , Calcium/metabolism , Cell Movement , Cell Proliferation , Central Nervous System/cytology , Central Nervous System/metabolism , Chemokines/classification , Chemokines/genetics , Humans , Microglia/metabolism , Neurons/metabolism , Receptors, Chemokine/metabolism , Synaptic Transmission/physiology
19.
Glia ; 54(8): 861-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16977602

ABSTRACT

Since activated microglia are able to phagocytose damaged cells and subsequently express major histocompatibility complex class II (MHC-II) and co-stimulatory proteins, they are considered to function as antigen presenting cells (APCs) in the central nervous system. The maturation and migratory potential of professional APCs is associated with the expression of chemokine receptor CCR7. We therefore investigated whether the immunological activation of microglia induces CCR7 expression. We here present that activation of cultured microglia by both the innate antigen lipopolysaccharide and protein antigen ovalbumin rapidly induces CCR7 expression, accompanied by increased MHC-II expression. Moreover, it is shown that CCR7 expression in IBA-1 positive cells is induced during the symptom onset and progression of experimental autoimmune encephalomyelitis, a rodent model for multiple sclerosis. These results suggest that microglia express CCR7 under specific inflammatory conditions, corroborating the idea that microglia develop into APCs with migratory potential toward lymphoid chemokines.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Chemotaxis/immunology , Gliosis/immunology , Microglia/immunology , Receptors, Chemokine/immunology , Animals , Animals, Newborn , Antigens/immunology , Disease Models, Animal , Encephalitis/immunology , Encephalitis/physiopathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Histocompatibility Antigens Class II/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Phagocytosis/immunology , Receptors, CCR7 , Receptors, Chemokine/genetics
20.
Neuropathol Appl Neurobiol ; 31(6): 600-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16281908

ABSTRACT

Feeding C57Bl/6 J mice the copper chelator cuprizone leads to selective apoptosis of mature oligodendrocytes and concomitant demyelination predominantly in the corpus callosum. The process of oligodendrocyte apoptosis in this animal model for multiple sclerosis (MS) involves early microglial activation, but no infiltration of T-lymphocytes. Therefore, this model could mimic early stages of oligodendrocyte degeneration Affected oligodendrocytes express the common neurotrophin receptor, p75(NTR), a 'stress-receptor' which under certain circumstances can induce apoptosis. Only affected oligodendrocytes in MS lesions and MS animal models express this receptor. In order to study the significance of p75(NTR) in the fate of oligodendrocytes, we have exposed wild-type as well as p75(NTR)-knockout mice to a 0.2% (w/w) cuprizone diet and performed a comparative immunohistochemical analysis of the corpus callosum at various time points. Surprisingly, our results show that the absence of p75(NTR) did not alter cuprizone-induced oligodendrocyte death (and subsequent de- or remyelination). Apparently, intracellular apoptosis pathways in adult oligodendrocytes do not require p75(NTR) activated signal transduction in the absence of T-lymphocytes and T-lymphocyte derived cytokines.


Subject(s)
Apoptosis , Demyelinating Diseases/pathology , Demyelinating Diseases/physiopathology , Oligodendroglia/pathology , Receptor, Nerve Growth Factor/genetics , Animals , Corpus Callosum/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoamine Oxidase Inhibitors , Nerve Regeneration , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...