Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Brain Behav Immun ; 119: 381-393, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38604270

ABSTRACT

INTRODUCTION: Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. Recent evidence suggests that lymphocyte trafficking in the intestines could play a key role in its etiology. Nevertheless, it is not clear how intestinal tissue is involved in the disease onset nor its evolution. In the present study, we aimed to evaluate intestinal inflammation dynamic throughout the disease course and its potential impact on disease progression. METHODS: We used tissue immunophenotyping (immunohistofluorescence and flow cytometry) and a recently described molecular magnetic resonance imaging (MRI) method targeting mucosal addressin cell adhesion molecule-1 (MAdCAM-1) to assess intestinal inflammation in vivo in two distinct animal models of MS (Experimental Autoimmune Encephalomyelitis - EAE) at several time points of disease progression. RESULTS: We report a positive correlation between disease severity and MAdCAM-1 MRI signal in two EAE models. Moreover, high MAdCAM-1 MRI signal during the asymptomatic phase is associated with a delayed disease onset in progressive EAE and to a lower risk of conversion to a secondary-progressive form in relapsing-remitting EAE. During disease evolution, in line with a bi-directional immune communication between the gut and the central nervous system, we observed a decrease in T-CD4+ and B lymphocytes in the ileum concomitantly with their increase in the spinal cord. CONCLUSION: Altogether, these data unveil a crosstalk between intestinal and central inflammation in EAE and support the use of molecular MRI of intestinal MAdCAM-1 as a new biomarker for prognostic in MS patients.

3.
J Inflamm (Lond) ; 21(1): 4, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38355547

ABSTRACT

Tissue-plasminogen activator (tPA) is a serine protease well known for its fibrinolytic function. Recent studies indicate that tPA could also modulate inflammation via plasmin generation and/or by receptor mediated signalling in vitro. However, the contribution of tPA in inflammatory processes in vivo has not been fully addressed. Therefore, using tPA-deficient mice, we have analysed the effect of lipopolysaccharide (LPS) challenge on the phenotype of myeloid cells including neutrophils, macrophages and dendritic cells (DCs) in spleen. We found that LPS treatment upregulated the frequency of major histocompatibility class two (MHCII+) macrophages but also, paradoxically, induced a deep downregulation of MHCII molecule level on macrophages and on conventional dendritic cells 2 (cDC2). Expression level of the CD11b integrin, known as a tPA receptor, was upregulated by LPS on MHCII+ macrophages and cDC2, suggesting that tPA effects could be amplified during inflammation. In tPA-/- mice under inflammatory conditions, expression of costimulatory CD86 molecules on MHCII+ macrophages was decreased compared to WT mice, while in steady state the expression of MHCII molecules was higher on macrophages. Finally, we reported that tPA deficiency slightly modified the phenotype of DCs and T cells in acute inflammatory conditions. Overall, our findings indicate that in vivo, LPS injection had an unexpectedly bimodal effect on MHCII expression on macrophages and DCs that consequently might affect adaptive immunity. tPA could also participate in the regulation of the T cell response by modulating the levels of CD86 and MHCII molecules on macrophages.

4.
Sante Publique ; 35(HS2): 107-109, 2024.
Article in French | MEDLINE | ID: mdl-38360765

ABSTRACT

The Groupe de réflexion avec les associations de malades (GRAM)—a joint scientist/association think tank at the French National Institute of Health and Medical Research (INSERM)—has reflected, on the basis of internal surveys and the experiences of its members, on focus points concerning good practice to help partners involved in participatory research projects. In this article, we list these focus points and highlight the need to afford partners opportunities for dialogue, in order to give good chances of success to participatory research projects, in both the interest of associations and researchers.


Subject(s)
Community-Based Participatory Research , Humans , Surveys and Questionnaires
5.
Cell Mol Life Sci ; 79(6): 323, 2022 May 28.
Article in English | MEDLINE | ID: mdl-35633384

ABSTRACT

BACKGROUND: In multiple sclerosis (MS), disturbance of the plasminogen activation system (PAS) and blood brain barrier (BBB) disruption are physiopathological processes that might lead to an abnormal fibrin(ogen) extravasation into the parenchyma. Fibrin(ogen) deposits, usually degraded by the PAS, promote an autoimmune response and subsequent demyelination. However, the PAS disruption is not well understood and not fully characterized in this disorder. METHODS: Here, we characterized the expression of PAS actors during different stages of two mouse models of MS (experimental autoimmune encephalomyelitis-EAE), in the central nervous system (CNS) by quantitative RT-PCR, immunohistofluorescence and fluorescent in situ hybridization (FISH). Thanks to constitutive PAI-1 knockout mice (PAI-1 KO) and an immunotherapy using a blocking PAI-1 antibody, we evaluated the role of PAI-1 in EAE models and its impact on physiopathological processes such as fibrin(ogen) deposits, lymphocyte infiltration and demyelination. RESULTS: We report a striking overexpression of PAI-1 in reactive astrocytes during symptomatic phases, in two EAE mouse models of MS. This increase is concomitant with lymphocyte infiltration and fibrin(ogen) deposits in CNS parenchyma. By genetic invalidation of PAI-1 in mice and immunotherapy using a blocking PAI-1 antibody, we demonstrate that abolition of PAI-1 reduces the severity of EAE and occurrence of relapses in two EAE models. These benefits are correlated with a decrease in fibrin(ogen) deposits, infiltration of T4 lymphocytes, reactive astrogliosis, demyelination and axonal damage. CONCLUSION: These results demonstrate that a deleterious overexpression of PAI-1 by reactive astrocytes leads to intra-parenchymal dysfibrinolysis in MS models and anti-PAI-1 strategies could be a new therapeutic perspective for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Plasminogen Activator Inhibitor 1 , Animals , Astrocytes/metabolism , Central Nervous System/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Fibrin , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Plasminogen Activator Inhibitor 1/genetics , Serpin E2
6.
Cell Immunol ; 371: 104451, 2022 01.
Article in English | MEDLINE | ID: mdl-34781155

ABSTRACT

The COVID-19 pandemic has once again brought to the forefront the existence of a tight link between the coagulation/fibrinolytic system and the immunologic processes. Tissue-type plasminogen activator (tPA) is a serine protease with a key role in fibrinolysis by converting plasminogen into plasmin that can finally degrade fibrin clots. tPA is released in the blood by endothelial cells and hepatocytes but is also produced by various types of immune cells including T cells and monocytes. Beyond its role on hemostasis, tPA is also a potent modulator of inflammation and is involved in the regulation of several inflammatory diseases. Here, after a brief description of tPA structure, we review its new functions in adaptive immunity focusing on T cells and antigen presenting cells. We intend to synthesize the recent knowledge on proteolysis- and receptor-mediated effects of tPA on immune response in physiological and pathological context.


Subject(s)
Blood Coagulation/immunology , COVID-19/immunology , Fibrinolysis/immunology , Immunity/immunology , SARS-CoV-2/immunology , Tissue Plasminogen Activator/immunology , Antigen-Presenting Cells/immunology , COVID-19/epidemiology , COVID-19/virology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Humans , Models, Immunological , Pandemics , SARS-CoV-2/physiology , T-Lymphocytes/immunology , Tissue Plasminogen Activator/metabolism
7.
J Thromb Haemost ; 19(9): 2235-2247, 2021 09.
Article in English | MEDLINE | ID: mdl-34060720

ABSTRACT

BACKGROUND: Factor XII (FXII) is a serine protease that participates in the intrinsic coagulation pathway. Several studies have shown that plasma FXII exerts a deleterious role in cerebral ischemia and traumatic brain injury by promoting thrombo-inflammation. Nevertheless, the impact of FXII on neuronal cell fate remains unknown. OBJECTIVES: We investigated the role of FXII and FXIIa in neuronal injury and apoptotic cell death. METHODS: We tested the neuroprotective roles of FXII and FXIIa in an experimental model of neuronal injury induced by stereotaxic intracerebral injection of N-methyl-D-aspartic acid (NMDA) in vivo and in a model of apoptotic death of murine primary neuronal cultures through serum deprivation in vitro. RESULTS: Here, we found that exogenous FXII and FXIIa reduce brain lesions induced by NMDA injection in vivo. Furthermore, FXII protects cultured neurons from apoptosis through a growth factor--like effect. This mechanism was triggered by direct interaction with epidermal growth factor (EGF) receptor and subsequent activation of this receptor. Interestingly, the "proteolytically" active and two-chain form of FXII, FXIIa, exerts its protective effects by an alternative signaling pathway. FXIIa activates the pro-form of hepatocyte growth factor (HGF) into HGF, which in turn activated the HGF receptor (HGFR) pathway. CONCLUSION: This study describes two novel mechanisms of action of FXII and identifies neurons as target cells for the protective effects of single and two-chain forms of FXII. Therefore, inhibition of FXII in neurological disorders may have deleterious effects by preventing its beneficial effects on neuronal survival.


Subject(s)
Factor XII , Proto-Oncogene Proteins c-met , Animals , Apoptosis , Blood Coagulation , Factor XIIa , Mice , Neurons
8.
J Neuroinflammation ; 18(1): 52, 2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33610187

ABSTRACT

BACKGROUND: Tissue plasminogen activator (tPA) is a serine protease involved in fibrinolysis. It is released by endothelial cells, but also expressed by neurons and glial cells in the central nervous system (CNS). Interestingly, this enzyme also contributes to pathological processes in the CNS such as neuroinflammation by activating microglia and increasing blood-brain barrier permeability. Nevertheless, its role in the control of adaptive and innate immune response remains poorly understood. METHODS: tPA effects on myeloid and lymphoid cell response were studied in vivo in the mouse model of multiple sclerosis experimental autoimmune encephalomyelitis and in vitro in splenocytes. RESULTS: tPA-/- animals exhibited less severe experimental autoimmune encephalomyelitis than their wild-type counterparts. This was accompanied by a reduction in both lymphoid and myeloid cell populations in the spinal cord parenchyma. In parallel, tPA increased T cell activation and proliferation, as well as cytokine production by a protease-dependent mechanism and via plasmin generation. In addition, tPA directly raised the expression of MHC-II and the co-stimulatory molecules CD80 and CD86 at the surface of dendritic cells and macrophages by a direct action dependent of the activation of epidermal growth factor receptor. CONCLUSIONS: Our study provides new insights into the mechanisms responsible for the harmful functions of tPA in multiple sclerosis and its animal models: tPA promotes the proliferation and activation of both lymphoid and myeloid populations by distinct, though complementary, mechanisms.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Lymphocyte Activation/drug effects , Myeloid Cells/drug effects , Tissue Plasminogen Activator/toxicity , Animals , Female , Humans , Lymphocyte Activation/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism , Tissue Plasminogen Activator/deficiency
9.
Mult Scler J Exp Transl Clin ; 6(4): 2055217320959806, 2020.
Article in English | MEDLINE | ID: mdl-33101703

ABSTRACT

BACKGROUND: Clinical observations support the hypothesis that stressful events increase relapse occurrence in multiple sclerosis patients, while stress-reduction strategies can modulate this effect. However, a direct cause-effect relationship between stress level and relapse cannot be firmly established from these data. OBJECTIVES: The purpose of this work was to address whether modulation of stress could interfere with symptom relapse in an animal model of multiple sclerosis with relapsing-remitting course. METHODS: Mice bred in standard or enriched environment were subjected to repeated acute stress during the remission phase of relapsing-remitting PLP-induced experimental autoimmune encephalomyelitis. RESULTS: We report that repeated acute stress induced a twofold increase in relapse incidence in experimental autoimmune encephalomyelitis. On the other hand, environmental enrichment reduced relapse incidence and severity, and reversed the effects of repeated acute stress. CONCLUSION: These data provide the platform for further studies on the biological processes that link stress and multiple sclerosis relapses in a suitable animal model.

10.
Brain ; 143(10): 2957-2972, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32893288

ABSTRACT

Anti-N-methyl-d-aspartate receptor (NMDAR) encephalitis is a neuropsychiatric disease characterized by an antibody-mediated autoimmune response against NMDAR. Recent studies have shown that anti-NMDAR antibodies are involved in the pathophysiology of the disease. However, the upstream immune and inflammatory processes responsible for this pathogenic response are still poorly understood. Here, we immunized mice against the region of NMDA receptor containing the N368/G369 amino acids, previously implicated in a pathogenic response. This paradigm induced encephalopathy characterized by blood-brain barrier opening, periventricular T2-MRI hyperintensities and IgG deposits into the brain parenchyma. Two weeks after immunization, mice developed clinical symptoms reminiscent of encephalitis: anxiety- and depressive-like behaviours, spatial memory impairment (without motor disorders) and increased sensitivity to seizures. This response occurred independently of overt T-cell recruitment. However, it was associated with B220+ (B cell) infiltration towards the ventricles, where they differentiated into CD138+ cells (plasmocytes). Interestingly, these B cells originated from peripheral lymphoid organs (spleen and cervical lymphoid nodes). Finally, blocking the B-cell response using a depleting cocktail of antibodies reduced the severity of symptoms in encephalitis mice. This study demonstrates that the B-cell response can lead to an autoimmune reaction against NMDAR that drives encephalitis-like behavioural impairments. It also provides a relevant platform for dissecting encephalitogenic mechanisms in an animal model, and enables the testing of therapeutic strategies targeting the immune system in anti-NMDAR encephalitis.


Subject(s)
Autoantibodies/blood , B-Lymphocytes/metabolism , Encephalitis/blood , Hashimoto Disease/blood , Nerve Tissue Proteins/toxicity , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , Encephalitis/chemically induced , Encephalitis/immunology , Hashimoto Disease/chemically induced , Hashimoto Disease/immunology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/immunology , Receptors, N-Methyl-D-Aspartate/immunology
11.
Sci Transl Med ; 12(560)2020 09 09.
Article in English | MEDLINE | ID: mdl-32908005

ABSTRACT

Mucosal tissues play critical roles in health and disease as the primary barrier between the external world and the inner body, lining the digestive, respiratory, urinary, mammary, and reproductive tracts. Clinical evaluation of mucosal tissues is currently performed using endoscopy, such as ileocolonoscopy for the intestinal mucosa, which causes substantial patient discomfort and can lead to organ damage. Here, we developed a contrast agent for molecular magnetic resonance imaging (MRI) that is targeted to mucosal vascular addressin cell adhesion molecule 1 (MAdCAM-1), an adhesion molecule overexpressed by inflamed mucosal tissues. We investigated the diagnostic performance of molecular MRI of MAdCAM-1 to detect mucosal inflammation in several models of acute and chronic intestinal inflammation in mice. We demonstrated that molecular MRI of MAdCAM-1 reveals disease activity and can evaluate the response to inflammatory treatments along the whole intestinal mucosa in clinically relevant models of inflammatory bowel diseases. We also provide evidence that this technique can detect low, subclinical mucosal inflammation. Molecular MRI of MAdCAM-1 has potential applications in early diagnosis, longitudinal follow-up, and therapeutic response monitoring in diseases affecting mucosal tissues, such as inflammatory bowel diseases.


Subject(s)
Immunoglobulins , Mucoproteins , Animals , Humans , Inflammation , Intestinal Mucosa , Leukocytes , Mice , Molecular Imaging
12.
J Neurosci ; 40(8): 1778-1787, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31953371

ABSTRACT

Increase in blood-brain barrier (BBB) permeability is a crucial step in neuroinflammatory processes. We previously showed that N Methyl D Aspartate Receptor (NMDARs), expressed on cerebral endothelial cells forming the BBB, regulate immune cell infiltration across this barrier in the mouse. Here, we describe the mechanism responsible for the action of NMDARs on BBB permeabilization. We report that mouse CNS endothelial NMDARs display the regulatory GluN3A subunit. This composition confers to NMDARs' unconventional properties: these receptors do not induce Ca2+ influx but rather show nonionotropic properties. In inflammatory conditions, costimulation of human brain endothelial cells by NMDA agonists (NMDA or glycine) and the serine protease tissue plasminogen activator, previously shown to potentiate NMDAR activity, induces metabotropic signaling via the Rho/ROCK pathway. This pathway leads to an increase in permeability via phosphorylation of myosin light chain and subsequent shrinkage of human brain endothelial cells. Together, these data draw a link between NMDARs and the cytoskeleton in brain endothelial cells that regulates BBB permeability in inflammatory conditions.SIGNIFICANCE STATEMENT The authors describe how NMDARs expressed on endothelial cells regulate blood-brain barrier function via myosin light chain phosphorylation and increase in permeability. They report that these non-neuronal NMDARs display distinct structural, functional, and pharmacological features than their neuronal counterparts.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Myosins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Blood-Brain Barrier/drug effects , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Endothelial Cells/drug effects , Excitatory Amino Acid Agonists/pharmacology , Male , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Permeability , Phosphorylation/drug effects , Receptors, N-Methyl-D-Aspartate/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Plasminogen Activator/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
13.
Front Immunol ; 10: 1081, 2019.
Article in English | MEDLINE | ID: mdl-31156634

ABSTRACT

CD4+ T cells differentiate into various T helper subsets characterized by distinct cytokine secreting profiles that confer them effector functions adapted to a variety of infectious or endogenous threats. Regulatory CD4+ T cells are another specialized subset that plays a fundamental role in the maintenance of immune tolerance to self-antigens. Manipulating effector or regulatory CD4+ T cells responses is a promising immunotherapy strategy for, respectively, chronical viral infections and cancer, or severe autoimmune diseases and transplantation. Adoptive cell therapy (ACT) is an emerging approach that necessitates defining robust and efficient methods for the in vitro expansion of antigen-specific T cells then infused into patients. To address this challenge, artificial antigen presenting cells (AAPCs) have been developed. They constitute a reliable and easily usable platform to stimulate and amplify antigen-specific CD4+ T cells. Here, we review the recent advances in understanding the functions of CD4+ T cells in immunity and in immune tolerance, and their use for ACT. We also describe the characteristics of different AAPC models and the way to improve their stimulating functions. Finally, we discuss the potential interest of these AAPCs, both as fundamental tools to decipher CD4+ T cell responses and as reagents to generate clinical grade antigen-specific CD4+ T cells for immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , T-Lymphocytes, Regulatory/immunology , Antigen Presentation , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , T-Lymphocytes, Regulatory/transplantation
14.
J Cereb Blood Flow Metab ; 39(7): 1258-1265, 2019 07.
Article in English | MEDLINE | ID: mdl-29376456

ABSTRACT

An alteration of parenchymal cerebrospinal fluid circulation (CSF) has been proposed to take part in the pathophysiology of multiple sclerosis. By using an intragate T1-weighted high-resolution MRI of the spinal cord of freely breathing mice injected with a gadolinium chelate in the cisterna magna, we show that a parenchymal CSF circulation exists in the spinal cord, in addition to that originally described in the brain. In experimental autoimmune encephalomyelitis, a model of multiple sclerosis, we show a reduction of parenchymal CSF circulation specifically in the spinal cord but not in the brain.


Subject(s)
Cerebrospinal Fluid/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Parenchymal Tissue/physiopathology , Spinal Cord/physiopathology , Animals , Brain/physiopathology , Cisterna Magna/drug effects , Contrast Media/administration & dosage , Female , Indocyanine Green/administration & dosage , Magnetic Resonance Imaging , Meglumine/administration & dosage , Mice , Multiple Sclerosis/physiopathology , Organometallic Compounds/administration & dosage
15.
Theranostics ; 8(5): 1195-1212, 2018.
Article in English | MEDLINE | ID: mdl-29507614

ABSTRACT

Endothelial cells of the central nervous system over-express surface proteins during neurological disorders, either as a cause, or a consequence, of the disease. Since the cerebral vasculature is easily accessible by large contrast-carrying particles, it constitutes a target of choice for molecular magnetic resonance imaging (MRI). In this review, we highlight the most recent advances in molecular MRI of brain endothelial activation and focus on the development of micro-sized particles of iron oxide (MPIO) targeting adhesion molecules including intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), P-Selectin and E-Selectin. We also discuss the perspectives and challenges for the clinical application of this technology in neurovascular disorders (ischemic stroke, intracranial hemorrhage, subarachnoid hemorrhage, diabetes mellitus), neuroinflammatory disorders (multiple sclerosis, brain infectious diseases, sepsis), neurodegenerative disorders (Alzheimer's disease, vascular dementia, aging) and brain cancers (primitive neoplasms, metastasis).


Subject(s)
Central Nervous System/pathology , Endothelial Cells/metabolism , Magnetic Resonance Imaging , Molecular Imaging , Animals , Central Nervous System/blood supply , Dextrans/chemistry , Endothelial Cells/pathology , Humans , Magnetite Nanoparticles/chemistry , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/pathology
16.
Glia ; 65(12): 1961-1975, 2017 12.
Article in English | MEDLINE | ID: mdl-28850711

ABSTRACT

Myelination is a late developmental process regulated by a set of inhibitory and stimulatory factors, including extracellular matrix components. Accordingly, chondroitin sulfate proteoglycans (CSPGs) act as negative regulators of myelination processes. A disintegrin and metalloproteinase with thrombospondin motifs type 4 (ADAMTS-4) is an extracellular protease capable of degrading CSPGs. Although exogenous ADAMTS-4 has been proven to be beneficial in several models of central nervous system (CNS) injuries, the physiological functions of endogenous ADAMTS-4 remain poorly understood. We first used Adamts4/LacZ reporter mice to reveal that ADAMTS-4 is strongly expressed in the CNS, especially in the white matter, with a cellular profile restricted to mature oligodendrocytes. Interestingly, we evidenced an abnormal myelination in Adamts4-/- mice, characterized by a higher diameter of myelinated axons with a shifting g-ratio. Accordingly, lack of ADAMTS-4 is accompanied by motor deficits and disturbed nervous electrical activity. In conclusion, we demonstrate that ADAMTS-4 is a new marker of mature oligodendrocytes contributing to the myelination processes and thus to the control of motor capacities.


Subject(s)
ADAMTS4 Protein/metabolism , Movement Disorders/genetics , Oligodendroglia/metabolism , ADAMTS4 Protein/genetics , Animals , Animals, Newborn , Calcium-Binding Proteins/metabolism , Corpus Callosum/metabolism , Corpus Callosum/pathology , Corpus Callosum/ultrastructure , Disease Models, Animal , Evoked Potentials, Somatosensory/genetics , Evoked Potentials, Somatosensory/physiology , Gait Disorders, Neurologic/etiology , Locomotion/genetics , Locomotion/physiology , Male , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Electron , Movement Disorders/physiopathology , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Statistics, Nonparametric , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
17.
Proc Natl Acad Sci U S A ; 114(23): 6116-6121, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28533365

ABSTRACT

New strategies for detecting disease activity in multiple sclerosis are being investigated to ameliorate diagnosis and follow-up of patients. Today, although magnetic resonance imaging (MRI) is widely used to diagnose and monitor multiple sclerosis, no imaging tools exist to predict the evolution of disease and the efficacy of therapeutic strategies. Here, we show that molecular MRI targeting the endothelial adhesion molecule P-selectin unmasks the pathological events that take place in the spinal cord of mice subjected to chronic or relapsing experimental autoimmune encephalomyelitis. This approach provides a quantitative spatiotemporal follow-up of disease course in relation to clinical manifestations. Moreover, it predicts relapse in asymptomatic mice and remission in symptomatic animals. Future molecular MRI targeting P-selectin may be used to improve diagnosis, follow-up of treatment, and management of relapse/remission cycles in multiple sclerosis patients by providing information currently inaccessible through conventional MRI techniques.


Subject(s)
Magnetic Resonance Imaging/methods , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Animals , Blood-Brain Barrier/diagnostic imaging , Brain/pathology , Contrast Media , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , P-Selectin/metabolism , Recurrence , Spinal Cord/pathology
18.
Cell Discov ; 3: 17001, 2017.
Article in English | MEDLINE | ID: mdl-28417010

ABSTRACT

Plasminogen activation is involved in many processes within the central nervous system, including synaptic plasticity, neuroinflammation and neurodegeneration. However, the mechanisms that regulate plasminogen activation in the brain still remain unknown. Here we demonstrate that astrocytes participate in this regulation by two mechanisms. First, the astrocyte plasma membrane serves as a surface for plasminogen activation by tissue-type plasminogen activator. This activation triggers downstream plasmin-dependent processes with important impacts in brain health and disease, such as fibrinolysis and brain-derived neurotrophic factor conversion. Second, astrocytes take up plasminogen and plasmin in a regulated manner through a novel mechanism involving endocytosis mediated by cell-surface actin and triggered by extracellular plasmin activity at the surface of astrocytes. Following endocytosis, plasminogen and plasmin are targeted to lysosomes for degradation. Thus, cell-surface actin acts as a sensor of plasmin activity to induce a negative feedback through plasmin endocytosis. This study provides evidence that astrocytes control the balance between plasmin formation and plasmin elimination in the brain parenchyma.

19.
Mol Neurodegener ; 12(1): 20, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28231842

ABSTRACT

BACKGROUND: The ability of oligodendrocyte progenitor cells (OPCs) to give raise to myelin forming cells during developmental myelination, normal adult physiology and post-lesion remyelination in white matter depends on factors which govern their proliferation, migration and differentiation. Tissue plasminogen activator (tPA) is a serine protease expressed in the central nervous system (CNS), where it regulates cell fate. In particular, tPA has been reported to protect oligodendrocytes from apoptosis and to facilitate the migration of neurons. Here, we investigated whether tPA can also participate in the migration of OPCs during CNS development and during remyelination after focal white matter lesion. METHODS: OPC migration was estimated by immunohistological analysis in spinal cord and corpus callosum during development in mice embryos (E13 to P0) and after white matter lesion induced by the stereotactic injection of lysolecithin in adult mice (1 to 21 days post injection). Migration was compared in these conditions between wild type and tPA knock-out animals. The action of tPA was further investigated in an in vitro chemokinesis assay. RESULTS: OPC migration along vessels is delayed in tPA knock-out mice during development and during remyelination. tPA enhances OPC migration via an effect dependent on the activation of epidermal growth factor receptor. CONCLUSION: Endogenous tPA facilitates the migration of OPCs during development and during remyelination after white matter lesion by the virtue of its epidermal growth factor-like domain.


Subject(s)
Cell Differentiation/drug effects , Central Nervous System/growth & development , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Tissue Plasminogen Activator/pharmacology , Animals , Brain Injuries/pathology , Cell Movement/drug effects , Central Nervous System/drug effects , Corpus Callosum/drug effects , Corpus Callosum/pathology , Embryo, Mammalian , Epidermal Growth Factor , Imaging, Three-Dimensional , Immunoblotting , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/drug effects , Neural Stem Cells/cytology , Oligodendroglia/cytology , White Matter/drug effects
20.
Lancet Neurol ; 15(10): 1089-102, 2016 09.
Article in English | MEDLINE | ID: mdl-27571160

ABSTRACT

Research advances support the idea that excessive activation of the glutamatergic pathway plays an important part in the pathophysiology of multiple sclerosis. Beyond the well established direct toxic effects on neurons, additional sites of glutamate-induced cell damage have been described, including effects in oligodendrocytes, astrocytes, endothelial cells, and immune cells. Such toxic effects could provide a link between various pathological aspects of multiple sclerosis, such as axonal damage, oligodendrocyte cell death, demyelination, autoimmunity, and blood-brain barrier dysfunction. Understanding of the mechanisms underlying glutamate toxicity in multiple sclerosis could help in the development of new approaches for diagnosis, treatment, and follow-up in patients with this debilitating disease. While several clinical trials of glutamatergic modulators have had disappointing results, our growing understanding suggests that there is reason to remain optimistic about the therapeutic potential of these drugs.


Subject(s)
Biomarkers/metabolism , Excitatory Amino Acid Antagonists/therapeutic use , Glutamic Acid/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...