Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 12167, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806485

ABSTRACT

During neuroinflammation, monocytes that infiltrate the central nervous system (CNS) may contribute to regenerative processes depending on their activation status. However, the extent and mechanisms of monocyte-induced CNS repair in patients with neuroinflammatory diseases remain largely unknown, partly due to the lack of a fully human assay platform that can recapitulate monocyte-neural stem cell interactions within the CNS microenvironment. We therefore developed a human model system to assess the impact of monocytic factors on neural stem cells, establishing a high-content compatible assay for screening monocyte-induced neural stem cell proliferation and differentiation. The model combined monocytes isolated from healthy donors and human embryonic stem cell derived neural stem cells and integrated both cell-intrinsic and -extrinsic properties. We identified CNS-mimicking culture media options that induced a monocytic phenotype resembling CNS infiltrating monocytes, while allowing adequate monocyte survival. Monocyte-induced proliferation, gliogenic fate and neurogenic fate of neural stem cells were affected by the conditions of monocytic priming and basal neural stem cell culture as extrinsic factors as well as the neural stem cell passage number as an intrinsic neural stem cell property. We developed a high-content compatible human in vitro assay for the integrated analysis of monocyte-derived factors on CNS repair.


Subject(s)
Cell Differentiation , Cell Proliferation , Monocytes , Neural Stem Cells , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/drug effects , Monocytes/cytology , Monocytes/metabolism , Monocytes/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Cells, Cultured
2.
Nat Commun ; 10(1): 217, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30644388

ABSTRACT

In demyelinating diseases including multiple sclerosis (MS), neural stem cells (NSCs) can replace damaged oligodendrocytes if the local microenvironment supports the required differentiation process. Although chitinase-like proteins (CLPs) form part of this microenvironment, their function in this differentiation process is unknown. Here, we demonstrate that murine Chitinase 3-like-3 (Chi3l3/Ym1), human Chi3L1 and Chit1 induce oligodendrogenesis. In mice, Chi3l3 is highly expressed in the subventricular zone, a stem cell niche of the adult brain, and in inflammatory brain lesions during experimental autoimmune encephalomyelitis (EAE). We find that silencing Chi3l3 increases severity of EAE. We present evidence that in NSCs Chi3l3 activates the epidermal growth factor receptor (EGFR), thereby inducing Pyk2-and Erk1/2- dependent expression of a pro-oligodendrogenic transcription factor signature. Our results implicate CLP-EGFR-Pyk2-MEK-ERK as a key intrinsic pathway controlling oligodendrogenesis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , ErbB Receptors/metabolism , Lectins/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , beta-N-Acetylhexosaminidases/metabolism , Animals , Chitinase-3-Like Protein 1/metabolism , Female , HEK293 Cells , Hexosaminidases/metabolism , Humans , MAP Kinase Signaling System , Mice
3.
Circ Res ; 117(9): 779-92, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26294656

ABSTRACT

RATIONALE: Platelets are known to participate in vascular pathologies; however, their role in neuroinflammatory diseases, such as multiple sclerosis (MS), is unknown. Autoimmune CD4 T cells have been the main focus of studies of MS, although the factors that regulate T-cell differentiation toward pathogenic T helper-1/T helper-17 phenotypes are not completely understood. OBJECTIVE: We investigated the role of platelets in the modulation of CD4 T-cell functions in patients with MS and in mice with experimental autoimmune encephalitis, an animal model for MS. METHODS AND RESULTS: We found that early in MS and experimental autoimmune encephalitis, platelets degranulated and produced soluble factors serotonin (5-hydroxytryptamine), platelet factor 4, and platelet-activating factor, which specifically stimulated differentiation of T cells toward pathogenic T helper-1, T helper-17, and interferon-γ/interleukin-17-producing CD4 T cells. At the later stages of MS and experimental autoimmune encephalitis, platelets became exhausted in their ability to produce proinflammatory factors and stimulate CD4 T cells but substantially increased their ability to form aggregates with CD4 T cells. Formation of platelet-CD4 T-cell aggregates involved the interaction of CD62P on activated platelets with adhesion molecule CD166 on activated CD4 T cells, contributing to downmodulation of CD4 T-cell activation, proliferation, and production of interferon-γ. Blocking of formation of platelet-CD4 T-cell aggregates during progression of experimental autoimmune encephalitis substantially enhanced proliferation of CD4 T cells in the central nervous system and the periphery leading to exacerbation of the disease. CONCLUSION: Our study indicates differential roles for platelets in the regulation of functions of pathogenic CD4 T cells during initiation and progression of central nervous system autoimmune inflammation.


Subject(s)
Blood Platelets/immunology , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Adult , Animals , Blood Platelets/metabolism , Blood Platelets/ultrastructure , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/ultrastructure , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Progression , Female , Flow Cytometry , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Middle Aged , Platelet Activating Factor/immunology , Platelet Activating Factor/metabolism , Platelet Factor 4/immunology , Platelet Factor 4/metabolism , Serotonin/immunology , Serotonin/metabolism
4.
Glycobiology ; 25(12): 1392-409, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26153105

ABSTRACT

Neural stem cell (NSC)-based therapies offer potential for neural repair in central nervous system (CNS) inflammatory and degenerative disorders. Typically, these conditions present with multifocal CNS lesions making it impractical to inject NSCs locally, thus mandating optimization of vascular delivery of the cells to involved sites. Here, we analyzed NSCs for expression of molecular effectors of cell migration and found that these cells are natively devoid of E-selectin ligands. Using glycosyltransferase-programmed stereosubstitution (GPS), we glycan engineered the cell surface of NSCs ("GPS-NSCs") with resultant enforced expression of the potent E-selectin ligand HCELL (hematopoietic cell E-/L-selectin ligand) and of an E-selectin-binding glycoform of neural cell adhesion molecule ("NCAM-E"). Following intravenous (i.v.) injection, short-term homing studies demonstrated that, compared with buffer-treated (control) NSCs, GPS-NSCs showed greater neurotropism. Administration of GPS-NSC significantly attenuated the clinical course of experimental autoimmune encephalomyelitis (EAE), with markedly decreased inflammation and improved oligodendroglial and axonal integrity, but without evidence of long-term stem cell engraftment. Notably, this effect of NSC is not a universal property of adult stem cells, as administration of GPS-engineered mouse hematopoietic stem/progenitor cells did not improve EAE clinical course. These findings highlight the utility of cell surface glycan engineering to boost stem cell delivery in neuroinflammatory conditions and indicate that, despite the use of a neural tissue-specific progenitor cell population, neural repair in EAE results from endogenous repair and not from direct, NSC-derived cell replacement.


Subject(s)
Cell Movement , Encephalomyelitis, Autoimmune, Experimental/therapy , Neural Stem Cells/metabolism , Polysaccharides/metabolism , Animals , Genetic Therapy , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred C57BL , Nerve Regeneration , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neural Stem Cells/transplantation , Selectins/metabolism
5.
PLoS One ; 9(5): e96256, 2014.
Article in English | MEDLINE | ID: mdl-24788965

ABSTRACT

BACKGROUND: Glatiramer acetate (GA, Copaxone, Copolymer-1) is an FDA approved drug for the treatment of MS and it is very effective in suppressing neuroinflammation in experimental autoimmune encephalitis (EAE), an animal model of MS. Although this drug was designed to inhibit pathogenic T cells, the exact mechanism of EAE/MS suppression by GA is still not well understood. Previously we presented evidence that platelets become activated and promote neuroinflammation in EAE, suggesting a possible pathogenic role of platelets in MS and EAE. We hypothesized that GA could inhibit neuroinflammation by affecting not only immune cells but also platelets. METHODOLOGY/PRINCIPAL FINDINGS: We investigated the effect of GA on the activation of human platelets in vitro: calcium influx, platelet aggregation and expression of activation markers. Our results in human platelets were confirmed by in-vitro and in-vivo studies of modulation of functions of platelets in mouse model. We found that GA inhibited thrombin-induced calcium influx in human and mouse platelets. GA also decreased thrombin-induced CD31, CD62P, CD63, and active form of αIIbß3 integrin surface expression and formation of platelet aggregates for both mouse and human platelets, and prolonged the bleeding time in mice by 2.7-fold. In addition, we found that GA decreased the extent of macrophage activation induced by co-culture of macrophages with platelets. CONCLUSIONS: GA inhibited the activation of platelets, which suggests a new mechanism of GA action in suppression of EAE/MS by targeting platelets and possibly preventing their interaction with immune cells such as macrophages. Furthermore, the reduction in platelet activation by GA may have additional cardiovascular benefits to prevent thrombosis.


Subject(s)
Calcium/metabolism , Peptides/pharmacology , Platelet Activation/drug effects , Thrombin/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , B7-2 Antigen/metabolism , Bleeding Time , Blood Platelets/drug effects , Blood Platelets/metabolism , Cells, Cultured , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Glatiramer Acetate , Histocompatibility Antigens Class II/metabolism , Humans , Ion Transport/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/prevention & control , P-Selectin/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
7.
PLoS One ; 8(3): e58979, 2013.
Article in English | MEDLINE | ID: mdl-23555611

ABSTRACT

Platelets respond to vascular damage and contribute to inflammation, but their role in the neurodegenerative diseases is unknown. We found that the systemic administration of brain lipid rafts induced a massive platelet activation and degranulation resulting in a life-threatening anaphylactic-like response in mice. Platelets were engaged by the sialated glycosphingolipids (gangliosides) integrated in the rigid structures of astroglial and neuronal lipid rafts. The brain-abundant gangliosides GT1b and GQ1b were specifically recognized by the platelets and this recognition involved multiple receptors with P-selectin (CD62P) playing the central role. During the neuroinflammation, platelets accumulated in the central nervous system parenchyma, acquired an activated phenotype and secreted proinflammatory factors, thereby triggering immune response cascades. This study determines a new role of platelets which directly recognize a neuronal damage and communicate with the cells of the immune system in the pathogenesis of neurodegenerative diseases.


Subject(s)
Blood Platelets/metabolism , Brain/metabolism , Glycolipids/metabolism , Membrane Microdomains/metabolism , Anaphylaxis/immunology , Anaphylaxis/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Biological Transport , Blood Platelets/immunology , Blood-Brain Barrier/metabolism , Brain/immunology , Cell Degranulation , Central Nervous System/immunology , Central Nervous System/metabolism , Cerebrovascular Disorders/immunology , Cerebrovascular Disorders/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gangliosides/immunology , Glycolipids/immunology , Inflammation/immunology , Inflammation/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/immunology , Mice , Neurons/immunology , Neurons/metabolism , Protein Binding , Receptors, Cell Surface/metabolism
8.
Immunity ; 37(2): 249-63, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22884314

ABSTRACT

Inflammation-mediated neurodegeneration occurs in the acute and the chronic phases of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Classically activated (M1) microglia are key players mediating this process. Here, we identified Galectin-1 (Gal1), an endogenous glycan-binding protein, as a pivotal regulator of M1 microglial activation that targets the activation of p38MAPK-, CREB-, and NF-κB-dependent signaling pathways and hierarchically suppresses downstream proinflammatory mediators, such as iNOS, TNF, and CCL2. Gal1 bound to core 2 O-glycans on CD45, favoring retention of this glycoprotein on the microglial cell surface and augmenting its phosphatase activity and inhibitory function. Gal1 was highly expressed in the acute phase of EAE, and its targeted deletion resulted in pronounced inflammation-induced neurodegeneration. Adoptive transfer of Gal1-secreting astrocytes or administration of recombinant Gal1 suppressed EAE through mechanisms involving microglial deactivation. Thus, Gal1-glycan interactions are essential in tempering microglial activation, brain inflammation, and neurodegeneration, with critical therapeutic implications for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Galectin 1/immunology , Leukocyte Common Antigens/metabolism , Microglia/immunology , Animals , Astrocytes/metabolism , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/physiopathology , Chemokine CCL2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Galectin 1/metabolism , Galectin 1/therapeutic use , Humans , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Polysaccharides/metabolism , Protein Binding , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
PLoS One ; 6(8): e23618, 2011.
Article in English | MEDLINE | ID: mdl-21886804

ABSTRACT

BACKGROUND: Type 1 regulatory T (Tr1) cells, characterized by the secretion of high levels of the anti-inflammatory cytokine interleukin-10 (IL-10), play an important role in the regulation of autoimmune diseases and transplantation. However, effective strategies that specifically induce Tr1 cells in vivo are limited. Furthermore, the pathways controlling the induction of these cells in vivo are not well understood. METHODOLOGY/PRINCIPAL FINDINGS: Here we report that nasal administration of anti-CD3 antibody induces suppressive Tr1 cells in mice. The in vivo induction of Tr1 cells by nasal anti-CD3 is dependent on IL-27 produced by upper airway resident dendritic cells (DCs), and is controlled by the transcription factors aryl hydrocarbon receptor (AHR) and c-Maf. Subsequently, IL-21 acts in an autocrine fashion to expand and maintain the Tr1 cells induced in vivo by nasally administered anti-CD3. CONCLUSIONS/SIGNIFICANCE: Our findings identify a unique approach to generate Tr1 cells in vivo and provide insights into the mechanisms by which these cells are induced.


Subject(s)
Dendritic Cells/immunology , Nasal Mucosa/immunology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Intranasal , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , Autoimmunity/drug effects , CD3 Complex/immunology , Dendritic Cells/drug effects , Interleukin-17/metabolism , Interleukins/metabolism , Mice , Models, Immunological , Nasal Mucosa/drug effects , Receptors, Interleukin-21/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/metabolism
10.
Ann Neurol ; 69(5): 878-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21391234

ABSTRACT

OBJECTIVE: The subventricular zone (SVZ) of the brain constitutes a niche for neural stem and progenitor cells that can initiate repair after central nervous system (CNS) injury. In a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE), the neural stem cells (NSCs) become activated and initiate regeneration during acute disease, but lose this ability during the chronic phases of disease. We hypothesized that chronic microglia activation contributes to the failure of the NSC repair potential in the SVZ. METHODS: Using bromodeoxyuridine injections at different time points during EAE, we quantified the number of proliferating and differentiating progenitors, and evaluated the structure of the SVZ by electron microscopy. In vivo minocycline treatment during EAE was used to address the effect of microglia inactivation on SVZ dysfunction. RESULTS: In vivo treatment with minocycline, an inhibitor of microglia activation, increases stem cell proliferation in both naive and EAE animals. Minocycline treatment decreases cortical and periventricular pathology in the chronic phase of EAE, improving the proliferation of Sox2 stem cells and NG2 oligodendrocyte precursors cells originating in the SVZ and their differentiation into mature oligodendrocytes. INTERPRETATION: These data suggest that failure of repair observed during chronic EAE correlates with microglia activation and that treatments targeting chronic microglial activation have the potential for enhancing repair in the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Neural Stem Cells/physiology , Stem Cell Niche/pathology , Animals , Anti-Bacterial Agents/pharmacology , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Mice , Microglia/drug effects , Microglia/pathology , Microglia/ultrastructure , Microscopy, Electron, Transmission , Minocycline/pharmacology , Multiple Sclerosis , Myelin Proteolipid Protein/adverse effects , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , Oligodendroglia/drug effects , Oligodendroglia/physiology , Peptide Fragments/adverse effects , Secondary Prevention , Stem Cell Niche/drug effects , Time Factors
11.
Brain Behav Immun ; 25(5): 991-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21074605

ABSTRACT

Microglia play an important role in inflammatory diseases of the central nervous system. There is evidence of microglial diversity with distinct phenotypes exhibiting either neuroprotection and repair or neurotoxicity. However the precise molecular mechanisms underlying this diversity are still unknown. Using a model of experimental autoimmune encephalomyelitis (EAE) we performed transcriptional profiling of isolated subventricular zone microglia from the acute and chronic disease phases of EAE. We found that microglia exhibit disease phase specific gene expression signatures, that correspond to unique gene ontology functions and genomic networks. Our data demonstrate for the first time, distinct transcriptional networks of microglia activation in vivo, that suggests a role as mediators of injury or repair.


Subject(s)
Gene Regulatory Networks/physiology , Microglia/physiology , Animals , Brain/cytology , Brain/metabolism , Brain/pathology , Brain/physiopathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Gene Expression Profiling , Gene Regulatory Networks/genetics , Mice , Microglia/metabolism , Microglia/pathology , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...