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










Database
Language
Publication year range
1.
Sci Rep ; 7(1): 16547, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29185463

ABSTRACT

IL-27 and IL-35 are heterodimeric cytokines, members of the IL-12 family and considered to have immunomodulatory properties. Their role during neuroinflammation had been investigated using mutant mice devoid of either one of their subunits or lacking components of their receptors, yielding conflicting results. We sought to understand the therapeutic potential of IL-27 and IL-35 delivered by gene therapy in neuroinflammation. We constructed lentiviral vectors expressing IL-27 and IL-35 from a single polypeptide chain, and we validated in vitro their biological activity. We injected IL-27 and IL-35-expressing lentiviral vectors into the cerebrospinal fluid (CSF) of mice affected by experimental neuroinflammation (EAE), and performed clinical, neuropathological and immunological analyses. Both cytokines interfere with neuroinflammation, but only IL-27 significantly modulates disease development, both clinically and neuropathologically. IL-27 protects from autoimmune inflammation by inhibiting granulocyte macrophages colony-stimulating factor (GM-CSF) expression in CD4+ T cells and by inducing program death-ligand 1 (PD-L1) expression in both CNS-resident and CNS-infiltrating myeloid cells. We demonstrate here that IL-27 holds therapeutic potential during neuroinflammation and that IL-27 inhibits GM-CSF and induces pd-l1 mRNA in vivo.


Subject(s)
Central Nervous System/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/metabolism , Interleukin-27/metabolism , Interleukins/metabolism , Leukocytes/metabolism , Animals , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cerebrospinal Fluid/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Genetic Therapy , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
2.
Neurobiol Aging ; 53: 159-168, 2017 05.
Article in English | MEDLINE | ID: mdl-28189343

ABSTRACT

Mutations in the TREM2 gene confer risk for Alzheimer's disease and susceptibility for Parkinson's disease (PD). We evaluated the effect of TREM2 deletion in a 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model, measuring neurodegeneration and microglia activation using a combined in vivo imaging and postmortem molecular approach. In wild-type mice, MPTP administration induced a progressive decrease of [11C]FECIT uptake, culminating at day 7. Neuronal loss was accompanied by an increase of TREM2, IL-1ß, and translocator protein (TSPO) transcript levels, [11C]PK11195 binding and GFAP staining (from day 2), and an early and transient increase of TNF-α, Galectin-3, and Iba-1 (from day 1). In TREM2 null (TREM2-/-) mice, MPTP similarly affected neuron viability and microglial cells, as shown by the lower level of Iba-1 staining in basal condition, and reduced increment of Iba-1, TNF-α, and IL-1ß in response to MPTP. Likely to compensate for TREM2 absence, TREM2-/- mice showed an earlier increment of [11C]PK11195 binding and a significant increase of IL-4. Taken together, our data demonstrate a central role of TREM2 in the regulation of microglia response to acute neurotoxic insults and suggest a potential modulatory role of TSPO in response to immune system deficit.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Deletion , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Nerve Degeneration/genetics , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Up-Regulation , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Acute Disease , Animals , Disease Models, Animal , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Nerve Degeneration/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/immunology
3.
J Neuroinflammation ; 13(1): 139, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27266518

ABSTRACT

BACKGROUND: Myeloid cells, such as macrophages and microglia, play a crucial role in neuroinflammation and have been recently identified as a novel therapeutic target, especially for chronic forms. The general aim would be to change the phenotype of myeloid cells from pro- to anti-inflammatory, favoring their tissue-trophic and regenerative functions. Myeloid cells, however, display a number of functional phenotypes, not immediately identifiable as pro- or anti-inflammatory, and associated to ambiguous markers. METHODS: We employed in vitro assays to study macrophage polarization/differentiation in the presence of classical polarizing stimuli such as IFNγ (pro-inflammatory) and IL4 (anti-inflammatory). We induced neuroinflammation in mice by immunization with a myelin antigen and treated diseased mice with intracisternal delivery of an IL4-expressing lentiviral vector. We analyzed clinical, pathological, and immunological outcomes with a focus on myeloid cells. RESULTS: We found that IL6, usually considered a pro-inflammatory cytokine, was released in vitro by macrophages treated with the anti-inflammatory cytokine IL4. We show the existence of macrophages expressing IL6 along with classical anti-inflammatory markers such as CD206 and demonstrate that these cells are immunosuppressive in vitro. In neuroinflamed mice, we show that IL4 delivery in the central nervous system (CNS) is associated with clinical and pathological protection from disease, associated with increased IL6 expression in infiltrating macrophages. CONCLUSIONS: IL6 is known to mediate both pro- and anti-inflammatory effects, having two distinct ways to induce cell-signaling: either through the membrane bound receptor (anti-inflammatory) or through trans-signaling (pro-inflammatory). We show here that IL6-expressing macrophages are associated to protection from neuroinflammation, suggesting that IL6 anti-inflammatory properties prevail in the CNS, and calling for a general reconsideration of IL6 in macrophage polarization.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-4/pharmacology , Interleukin-6/biosynthesis , Macrophages/metabolism , Animals , Cells, Cultured , Coculture Techniques , Dose-Response Relationship, Drug , Female , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Interleukin-4/immunology , Interleukin-6/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL
4.
J Biol Chem ; 288(42): 30210-30222, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24019529

ABSTRACT

Myeloid differentiation factor 88 (MyD88) is an adaptor protein that transduces intracellular signaling pathways evoked by the Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs). MyD88 is composed of an N-terminal death domain (DD) and a C-terminal Toll/IL-1 receptor (TIR) domain, separated by a short region. Upon ligand binding, TLR/IL-1Rs hetero- or homodimerize and recruit MyD88 through their respective TIR domains. Then, MyD88 oligomerizes via its DD and TIR domain and interacts with the interleukin-1 receptor-associated kinases (IRAKs) to form the Myddosome complex. We performed site-directed mutagenesis of conserved residues that are located in exposed regions of the MyD88-TIR domain and analyzed the effect of the mutations on MyD88 signaling. Our studies revealed that mutation of Glu(183), Ser(244), and Arg(288) impaired homodimerization of the MyD88-TIR domain, recruitment of IRAKs, and activation of NF-κB. Moreover, overexpression of two green fluorescent protein (GFP)-tagged MyD88 mini-proteins (GFP-MyD88151-189 and GFP-MyD88168-189), comprising the Glu(183) residue, recapitulated these effects. Importantly, expression of these dominant negative MyD88 mini-proteins competed with the function of endogenous MyD88 and interfered with TLR2/4-mediated responses in a human monocytic cell line (THP-1) and in human primary monocyte-derived dendritic cells. Thus, our studies identify novel residues of the TIR domain that are crucially involved in MyD88 homodimerization and TLR signaling in immune cells.


Subject(s)
Dendritic Cells/metabolism , Monocytes/metabolism , Myeloid Differentiation Factor 88/metabolism , Protein Multimerization/physiology , Signal Transduction/physiology , Amino Acid Substitution , Cell Line, Tumor , Dendritic Cells/cytology , Dendritic Cells/immunology , HEK293 Cells , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Monocytes/cytology , Monocytes/immunology , Mutagenesis, Site-Directed , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Protein Structure, Tertiary , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
5.
Ann Neurol ; 72(4): 610-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23109155

ABSTRACT

OBJECTIVE: Microvesicles (MVs) have been indicated as important mediators of intercellular communication and are emerging as new biomarkers of tissue damage. Our previous data indicate that reactive microglia/macrophages release MVs in vitro. The aim of the study was to evaluate whether MVs are released by microglia/macrophages in vivo and whether their number varies in brain inflammatory conditions, such as multiple sclerosis (MS). METHODS: Electron and fluorescence microscopy and flow cytometry were used to detect myeloid MVs in the cerebrospinal fluid (CSF) of healthy controls, MS patients, and rodents affected by experimental autoimmune encephalomyelitis (EAE), the animal model of MS. RESULTS: Myeloid MVs were detected in CSF of healthy controls. In relapsing and remitting EAE mice, the concentration of myeloid MVs in the CSF was significantly increased and closely associated with disease course. Analysis of MVs in the CSF of 28 relapsing patients and 28 patients with clinical isolated syndrome from 2 independent cohorts revealed higher levels of myeloid MVs than in 13 age-matched controls, indicating a clinical value of MVs as a companion tool to capture disease activity. Myeloid MVs were found to spread inflammatory signals both in vitro and in vivo at the site of administration; mice impaired in MV shedding were protected from EAE, suggesting a pathogenic role for MVs in the disease. Finally, FTY720, the first approved oral MS drug, significantly reduced the amount of MVs in the CSF of EAE-treated mice. INTERPRETATION: These findings identify myeloid MVs as a marker and therapeutic target of brain inflammation.


Subject(s)
Biomarkers/cerebrospinal fluid , Central Nervous System Diseases/cerebrospinal fluid , Central Nervous System Diseases/drug therapy , Inflammation/cerebrospinal fluid , Inflammation/drug therapy , Spinal Cord/metabolism , Animals , Blotting, Western , Calcium Signaling/physiology , Cell Communication , Cells, Cultured , Encephalitis/cerebrospinal fluid , Encephalitis/pathology , Flow Cytometry , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Multiple Sclerosis/pathology , Nervous System Autoimmune Disease, Experimental/cerebrospinal fluid , Nervous System Autoimmune Disease, Experimental/drug therapy , Neuroglia/metabolism , Neuroglia/physiology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...