Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
2.
EMBO J ; 43(8): 1420-1444, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38528182

ABSTRACT

Current approaches to the treatment of schizophrenia have mainly focused on the protein-coding part of the genome; in this context, the roles of microRNAs have received less attention. In the present study, we analyze the microRNAome in the blood and postmortem brains of schizophrenia patients, showing that the expression of miR-99b-5p is downregulated in both the prefrontal cortex and blood of patients. Lowering the amount of miR-99b-5p in mice leads to both schizophrenia-like phenotypes and inflammatory processes that are linked to synaptic pruning in microglia. The microglial miR-99b-5p-supressed inflammatory response requires Z-DNA binding protein 1 (Zbp1), which we identify as a novel miR-99b-5p target. Antisense oligonucleotides against Zbp1 ameliorate the pathological effects of miR-99b-5p inhibition. Our findings indicate that a novel miR-99b-5p-Zbp1 pathway in microglia might contribute to the pathogenesis of schizophrenia.


Subject(s)
MicroRNAs , Schizophrenia , Animals , Humans , Mice , Microglia/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Schizophrenia/genetics
3.
Biochim Biophys Acta Mol Cell Res ; 1870(6): 119485, 2023 08.
Article in English | MEDLINE | ID: mdl-37150482

ABSTRACT

Ca2+ signaling is one of the essential signaling systems for T lymphocyte activation, the latter being an essential step in the pathogenesis of autoimmune diseases such as multiple sclerosis (MS). Store-operated Ca2+ entry (SOCE) ensures long lasting Ca2+ signaling and is of utmost importance for major downstream T lymphocyte activation steps, e.g. nuclear localization of the transcription factor 'nuclear factor of activated T cells' (NFAT). 2-Methoxyestradiol (2ME2), an endogenous metabolite of estradiol (E2), blocks nuclear translocation of NFAT. The likely underlying mechanism is inhibition of SOCE, as shown for its synthetic sulfamate ester analogue 2-ethyl-3-sulfamoyloxy-17ß-cyanomethylestra-1,3,5(10)-triene (STX564). Here, we demonstrate that another synthetic bis-sulfamoylated 2ME2 derivative, 2-methoxyestradiol-3,17-O,O-bis-sulfamate (2-MeOE2bisMATE, STX140), an orally bioavailable, multi-targeting anticancer agent and potent steroid sulfatase (STS) inhibitor, antagonized SOCE in T lymphocytes. Downstream events, e.g. secretion of the pro-inflammatory cytokines interferon-γ and interleukin-17, were decreased by STX140 in in vitro experiments. Remarkably, STX140 dosed in vivo completely blocked the clinical disease in both active and transfer experimental autoimmune encephalomyelitis (EAE) in Lewis rats, a T cell-mediated animal model for MS, at a dose of 10 mg/kg/day i.p., whereas neither 2ME2 nor Irosustat, a pure STS inhibitor, showed any effect. The STS inhibitory activity of STX140 is therefore not responsible for its activity in this model. Taken together, inhibition of SOCE by STX140 resulting in full antagonism of clinical symptoms in EAE in the Lewis rat, paired with the known excellent bioavailability and pharmaceutical profile of this drug, open potentially new therapeutic avenues for the treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , T-Lymphocytes , Rats , Animals , 2-Methoxyestradiol , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Rats, Inbred Lew , Pharmaceutical Preparations
4.
Sci Adv ; 8(37): eabo7639, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36112685

ABSTRACT

To maintain homeostasis, the body, including the brain, reprograms its metabolism in response to altered nutrition or disease. However, the consequences of these challenges for the energy metabolism of the different brain cell types remain unknown. Here, we generated a proteome atlas of the major central nervous system (CNS) cell types from young and adult mice, after feeding the therapeutically relevant low-carbohydrate, high-fat ketogenic diet (KD) and during neuroinflammation. Under steady-state conditions, CNS cell types prefer distinct modes of energy metabolism. Unexpectedly, the comparison with KD revealed distinct cell type-specific strategies to manage the altered availability of energy metabolites. Astrocytes and neurons but not oligodendrocytes demonstrated metabolic plasticity. Moreover, inflammatory demyelinating disease changed the neuronal metabolic signature in a similar direction as KD. Together, these findings highlight the importance of the metabolic cross-talk between CNS cells and between the periphery and the brain to manage altered nutrition and neurological disease.


Subject(s)
Brain , Diet, Ketogenic , Animals , Brain/metabolism , Carbohydrates , Ketone Bodies/metabolism , Mice , Proteome/metabolism
5.
Nat Neurosci ; 25(7): 887-899, 2022 07.
Article in English | MEDLINE | ID: mdl-35773544

ABSTRACT

The meninges, comprising the leptomeninges (pia and arachnoid layers) and the pachymeninx (dura layer), participate in central nervous system (CNS) autoimmunity, but their relative contributions remain unclear. Here we report on findings in animal models of CNS autoimmunity and in patients with multiple sclerosis, where, in acute and chronic disease, the leptomeninges were highly inflamed and showed structural changes, while the dura mater was only marginally affected. Although dural vessels were leakier than leptomeningeal vessels, effector T cells adhered more weakly to the dural endothelium. Furthermore, local antigen-presenting cells presented myelin and neuronal autoantigens less efficiently, and the activation of autoreactive T cells was lower in dural than leptomeningeal layers, preventing local inflammatory processes. Direct antigen application was required to evoke a local inflammatory response in the dura. Together, our data demonstrate an uneven involvement of the meningeal layers in CNS autoimmunity, in which effector T cell trafficking and activation are functionally confined to the leptomeninges, while the dura remains largely excluded from CNS autoimmune processes.


Subject(s)
Autoimmunity , Meninges , Multiple Sclerosis , Animals , Arachnoid , Central Nervous System , Dura Mater , Humans , Meninges/physiology
6.
Nature ; 603(7899): 138-144, 2022 03.
Article in English | MEDLINE | ID: mdl-35197636

ABSTRACT

Lung infections and smoking are risk factors for multiple sclerosis, a T-cell-mediated autoimmune disease of the central nervous system1. In addition, the lung serves as a niche for the disease-inducing T cells for long-term survival and for maturation into migration-competent effector T cells2. Why the lung tissue in particular has such an important role in an autoimmune disease of the brain is not yet known. Here we detected a tight interconnection between the lung microbiota and the immune reactivity of the brain. A dysregulation in the lung microbiome significantly influenced the susceptibility of rats to developing autoimmune disease of the central nervous system. Shifting the microbiota towards lipopolysaccharide-enriched phyla by local treatment with neomycin induced a type-I-interferon-primed state in brain-resident microglial cells. Their responsiveness towards autoimmune-dominated stimulation by type II interferons was impaired, which led to decreased proinflammatory response, immune cell recruitment and clinical signs. Suppressing lipopolysaccharide-producing lung phyla with polymyxin B led to disease aggravation, whereas addition of lipopolysaccharide-enriched phyla or lipopolysaccharide recapitulated the neomycin effect. Our data demonstrate the existence of a lung-brain axis in which the pulmonary microbiome regulates the immune reactivity of the central nervous tissue and thereby influences its susceptibility to autoimmune disease development.


Subject(s)
Autoimmunity , Brain , Microbiota , Multiple Sclerosis , Animals , Autoimmune Diseases , Brain/physiology , Lipopolysaccharides/pharmacology , Lung/microbiology , Multiple Sclerosis/etiology , Neomycin , Rats
7.
Cell Rep ; 37(4): 109889, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706227

ABSTRACT

Astrocyte-derived cholesterol supports brain cells under physiological conditions. However, in demyelinating lesions, astrocytes downregulate cholesterol synthesis, and the cholesterol that is essential for remyelination has to originate from other cellular sources. Here, we show that repair following acute versus chronic demyelination involves distinct processes. In particular, in chronic myelin disease, when recycling of lipids is often defective, de novo neuronal cholesterol synthesis is critical for regeneration. By gene expression profiling, genetic loss-of-function experiments, and comprehensive phenotyping, we provide evidence that neurons increase cholesterol synthesis in chronic myelin disease models and in patients with multiple sclerosis (MS). In mouse models, neuronal cholesterol facilitates remyelination specifically by triggering oligodendrocyte precursor cell proliferation. Our data contribute to the understanding of disease progression and have implications for therapeutic strategies in patients with MS.


Subject(s)
Cholesterol , Multiple Sclerosis , Myelin Sheath , Oligodendrocyte Precursor Cells/metabolism , Remyelination/genetics , Animals , Cholesterol/biosynthesis , Cholesterol/genetics , Disease Models, Animal , Humans , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism
8.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33645550

ABSTRACT

Approximately 80% of neuromyelitis optica spectrum disorder (NMOSD) patients harbor serum anti-aquaporin-4 autoantibodies targeting astrocytes in the CNS. Crucial for NMOSD lesion initiation is disruption of the blood-brain barrier (BBB), which allows the entrance of Abs and serum complement into the CNS and which is a target for new NMOSD therapies. Astrocytes have important functions in BBB maintenance; however, the influence of their loss and the role of immune cell infiltration on BBB permeability in NMOSD have not yet been investigated. Using an experimental model of targeted NMOSD lesions in rats, we demonstrate that astrocyte destruction coincides with a transient disruption of the BBB and a selective loss of occludin from tight junctions. It is noteworthy that BBB integrity is reestablished before astrocytes repopulate. Rather than persistent astrocyte loss, polymorphonuclear leukocytes (PMNs) are the main mediators of BBB disruption, and their depletion preserves BBB integrity and prevents astrocyte loss. Inhibition of PMN chemoattraction, activation, and proteolytic function reduces lesion size. In summary, our data support a crucial role for PMNs in BBB disruption and NMOSD lesion development, rendering their recruitment and activation promising therapeutic targets.


Subject(s)
Astrocytes/immunology , Blood-Brain Barrier/immunology , Leukocytes, Mononuclear/immunology , Neuromyelitis Optica/immunology , Animals , Astrocytes/pathology , Blood-Brain Barrier/pathology , Disease Models, Animal , Female , Humans , Leukocytes, Mononuclear/pathology , Neuromyelitis Optica/pathology , Rats , Rats, Inbred Lew
9.
Biochim Biophys Acta Mol Cell Res ; 1868(6): 118988, 2021 05.
Article in English | MEDLINE | ID: mdl-33581218

ABSTRACT

T cell activation starts with formation of second messengers that release Ca2+ from the endoplasmic reticulum (ER) and thereby activate store-operated Ca2+ entry (SOCE), one of the essential signals for T cell activation. Recently, the steroidal 2-methoxyestradiol was shown to inhibit nuclear translocation of the nuclear factor of activated T cells (NFAT). We therefore investigated 2-methoxyestradiol for inhibition of Ca2+ entry in T cells, screened a library of 2-methoxyestradiol analogues, and characterized the derivative 2-ethyl-3-sulfamoyloxy-17ß-cyanomethylestra-1,3,5(10)-triene (STX564) as a novel, potent and specific SOCE inhibitor. STX564 inhibits Ca2+ entry via SOCE without affecting other ion channels and pumps involved in Ca2+ signaling in T cells. Downstream effects such as cytokine expression and cell proliferation were also inhibited by both 2-methoxyestradiol and STX564, which has potential as a new chemical biology tool.


Subject(s)
2-Methoxyestradiol/pharmacology , Calcium Signaling/drug effects , Estrenes/pharmacology , NFATC Transcription Factors/metabolism , T-Lymphocytes/cytology , 2-Methoxyestradiol/analogs & derivatives , Animals , Calcium/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Estrenes/chemical synthesis , Estrenes/chemistry , Gene Expression Regulation/drug effects , Humans , Jurkat Cells , Lymphocyte Activation/drug effects , Protein Transport/drug effects , Rats , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
Nat Neurosci ; 24(1): 47-60, 2021 01.
Article in English | MEDLINE | ID: mdl-33349711

ABSTRACT

The repair of inflamed, demyelinated lesions as in multiple sclerosis (MS) necessitates the clearance of cholesterol-rich myelin debris by microglia/macrophages and the switch from a pro-inflammatory to an anti-inflammatory lesion environment. Subsequently, oligodendrocytes increase cholesterol levels as a prerequisite for synthesizing new myelin membranes. We hypothesized that lesion resolution is regulated by the fate of cholesterol from damaged myelin and oligodendroglial sterol synthesis. By integrating gene expression profiling, genetics and comprehensive phenotyping, we found that, paradoxically, sterol synthesis in myelin-phagocytosing microglia/macrophages determines the repair of acutely demyelinated lesions. Rather than producing cholesterol, microglia/macrophages synthesized desmosterol, the immediate cholesterol precursor. Desmosterol activated liver X receptor (LXR) signaling to resolve inflammation, creating a permissive environment for oligodendrocyte differentiation. Moreover, LXR target gene products facilitated the efflux of lipid and cholesterol from lipid-laden microglia/macrophages to support remyelination by oligodendrocytes. Consequently, pharmacological stimulation of sterol synthesis boosted the repair of demyelinated lesions, suggesting novel therapeutic strategies for myelin repair in MS.


Subject(s)
Demyelinating Diseases/pathology , Microglia/physiology , Sterols/biosynthesis , Animals , Cholesterol/metabolism , Desmosterol/metabolism , Encephalomyelitis, Autoimmune, Experimental , Female , Gene Expression Profiling , Humans , Inflammation/metabolism , Inflammation/pathology , Lipid Metabolism , Liver X Receptors/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis , Oligodendroglia/metabolism , Phagocytosis , Squalene/metabolism
11.
Nature ; 567(7749): E15, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30867589

ABSTRACT

In this Article, owing to an error during the production process, the y-axis label of Fig. 2c should read "Number of Tß-syn cells" rather than "Number of T1ß-syn cells" and the left and right panels of Fig. 4 should be labelled 'a' and 'b', respectively. These errors have been corrected online.

12.
Nature ; 566(7745): 503-508, 2019 02.
Article in English | MEDLINE | ID: mdl-30787438

ABSTRACT

The grey matter is a central target of pathological processes in neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. The grey matter is often also affected in multiple sclerosis, an autoimmune disease of the central nervous system. The mechanisms that underlie grey matter inflammation and degeneration in multiple sclerosis are not well understood. Here we show that, in Lewis rats, T cells directed against the neuronal protein ß-synuclein specifically invade the grey matter and that this is accompanied by the presentation of multifaceted clinical disease. The expression pattern of ß-synuclein induces the local activation of these T cells and, therefore, determined inflammatory priming of the tissue and targeted recruitment of immune cells. The resulting inflammation led to significant changes in the grey matter, which ranged from gliosis and neuronal destruction to brain atrophy. In humans, ß-synuclein-specific T cells were enriched in patients with chronic-progressive multiple sclerosis. These findings reveal a previously unrecognized role of ß-synuclein in provoking T-cell-mediated pathology of the central nervous system.


Subject(s)
Gray Matter/immunology , Gray Matter/pathology , Multiple Sclerosis, Chronic Progressive/immunology , Multiple Sclerosis, Chronic Progressive/pathology , T-Lymphocytes/immunology , beta-Synuclein/immunology , Animals , Brain/pathology , Cell Movement/immunology , Female , Gene Expression Regulation , Gliosis/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Lymphocyte Activation , Lymphocyte Count , Male , Multiple Sclerosis, Chronic Progressive/blood , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neurons/pathology , Rats , Rats, Inbred Lew , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , beta-Synuclein/analysis , beta-Synuclein/genetics , beta-Synuclein/metabolism
13.
Hum Mol Genet ; 28(1): 31-50, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30219847

ABSTRACT

Alpha-synuclein (aSyn) is a central player in Parkinson's disease (PD) but the precise molecular mechanisms underlying its pathogenicity remain unclear. It has recently been suggested that nuclear aSyn may modulate gene expression, possibly via interactions with DNA. However, the biological behavior of aSyn in the nucleus and the factors affecting its transcriptional role are not known. Here, we investigated the mechanisms underlying aSyn-mediated transcription deregulation by assessing its effects in the nucleus and the impact of phosphorylation in these dynamics. We found that aSyn induced severe transcriptional deregulation, including the downregulation of important cell cycle-related genes. Importantly, transcriptional deregulation was concomitant with reduced binding of aSyn to DNA. By forcing the nuclear presence of aSyn in the nucleus (aSyn-NLS), we found the accumulation of high molecular weight aSyn species altered gene expression and reduced toxicity when compared with the wild-type or exclusively cytosolic protein. Interestingly, nuclear localization of aSyn, and the effect on gene expression and cytotoxicity, was also modulated by phosphorylation on serine 129. Thus, we hypothesize that the role of aSyn on gene expression and, ultimately, toxicity, may be modulated by the phosphorylation status and nuclear presence of different aSyn species. Our findings shed new light onto the subcellular dynamics of aSyn and unveil an intricate interplay between subcellular location, phosphorylation and toxicity, opening novel avenues for the design of future strategies for therapeutic intervention in PD and other synucleinopathies.


Subject(s)
alpha-Synuclein/metabolism , alpha-Synuclein/physiology , Animals , Cell Line , Cell Nucleus , DNA-Binding Proteins , Down-Regulation , Gene Expression , Gene Expression Regulation/physiology , Humans , Mice , Nuclear Localization Signals/physiology , Parkinson Disease/pathology , Phosphorylation , Primary Cell Culture , Rats
14.
J Neuroinflammation ; 15(1): 217, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30068351

ABSTRACT

BACKGROUND: Autoimmune polyneuropathies are acquired inflammatory disorders of the peripheral nervous system (PNS) characterized by inflammation, demyelination, and axonal degeneration. Although the pathogenesis has not been fully elucidated, T cells recognizing self-antigens are believed to initiate inflammation in a subgroup of patients. However, the route and time of T cell entry into the PNS have not yet been described in detail. In this study, we analyzed both kinetics as well as localization of retrovirally transfected green fluorescent protein (GFP)-expressing neuritogenic T lymphocytes in experimental autoimmune neuritis (EAN). METHODS: T lymphocytes obtained from rats following EAN induction by immunization with peripheral nerve protein peptide P255-78 were retrovirally engineered to express GFP. Non-specific T cells were negatively selected by in vitro restimulation, whereas GFP-expressing neuritogenic T cells (reactive to P255-78) were adoptively transferred into healthy rats (AT-EAN). Antigen-specific T cell tracking and localization was performed by flow cytometry and immunohistochemistry during the course of disease. RESULTS: After induction of autoimmune neuritis, P2-reactive T cells were detectable in the liver, spleen, lymph nodes, lung, peripheral blood, and the sciatic nerves with distinct kinetics. A significant number of GFP+ T cells appeared early in the lung with a peak at day four. In the peripheral nerves within the first days, GFP-negative T cells rapidly accumulated and exceeded the number of GFP-expressing cells, but did not enter the endoneurium. Very early after adoptive transfer, T cells are found in proximity to peripheral nerves and in the epineurium. However, only GFP-expressing neuritogenic T cells are able to enter the endoneurium from day five after transfer. CONCLUSIONS: Our findings suggest that neuritogenic T cells invade the PNS early in the course of disease. However, neuritogenic T cells cross the blood-nerve barrier with a certain delay without preference to dorsal roots. Further understanding of the pathophysiological role of autoagressive T cells may help to improve therapeutic strategies in immune-mediated neuropathies.


Subject(s)
Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , Peripheral Nerves/pathology , T-Lymphocytes/physiology , Adoptive Transfer , Animals , CD4 Antigens/metabolism , Cell Proliferation/physiology , Disease Models, Animal , Female , Flow Cytometry , Freund's Adjuvant/toxicity , Gene Expression Regulation/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Myelin P2 Protein/metabolism , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/surgery , Peptide Fragments/metabolism , Rats , Rats, Inbred Lew , T-Lymphocytes/metabolism , Time Factors , Transduction, Genetic
15.
Mol Cell Neurosci ; 85: 105-118, 2017 12.
Article in English | MEDLINE | ID: mdl-28889992

ABSTRACT

Based on our previous demonstration of CXCR7 as the major mediator of CXCL12 signaling in cultured astrocytes, we have now compared astrocytic expression of the CXCL12 receptors, CXCR7 and CXCR4, during CNS development and disease. In addition, we asked whether disease-associated conditions/factors affect expression of CXCL12 receptors in astrocytes. In the late embryonic rat brain, CXCR7+/GFAP+ cells were restricted to the ventricular/subventricular zone while CXCR4 was widely absent from GFAP-positive cells. In the early postnatal and adult brain, CXCR7 and CXCR4 were almost exclusively expressed by GFAP-immunoreactive astrocytes forming the superficial glia limitans. Contrasting the situation in the intact CNS, a striking increase in astrocytic CXCR7 expression was detectable in the cortex of rats with experimental brain infarcts, in the spinal cord of rats with experimental autoimmune encephalomyelitis (EAE) and after mechanical compression, as well as in the in infarcted human cerebral cortex and in the hippocampus of Alzheimer's disease patients. None of these pathologies was associated with substantial increases in astrocytic CXCR4 expression. Screening of various disease-associated factors/conditions further revealed that CXCR7 expression of cultured cortical astrocytes increases with IFNγ as well as under hypoxic conditions whereas CXCR7 expression is attenuated following treatment with IFNß. Again, none of the treatments affected CXCR4 expression in cultured astrocytes. Together, these findings support the hypothesis of a crucial role of astrocytic CXCR7 in the progression of various CNS pathologies.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Central Nervous System Diseases/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR/biosynthesis , Aged , Animals , Brain/embryology , Brain/growth & development , Humans , Middle Aged , Rats , Rats, Sprague-Dawley
16.
Neurobiol Dis ; 102: 60-69, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28235673

ABSTRACT

Laquinimod is currently being tested as a therapeutic drug in multiple sclerosis. However, its exact mechanism of action is still under investigation. Tracking of fluorescently-tagged encephalitogenic T cells during experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, revealed that laquinimod significantly reduces the invasion of pathogenic effector T cells into the CNS tissue. T-cell activation, differentiation and amplification within secondary lymphoid organs after immunization with myelin antigen, their migratory capacity and re-activation within the nervous tissue were either only mildly affected or remained unchanged. Instead, laquinimod directly impacted the functionality of the CNS vasculature. The expression of tight junction proteins p120 and ZO-1 in human brain endothelial cells was up-regulated upon laquinimod treatment, resulting in a significant increase in the transendothelial electrical resistance of confluent monolayers of brain endothelial cells. Similarly, expression of the adhesion molecule activated leukocyte cell adhesion molecule (ALCAM) and inflammatory chemokines CCL2 and IP-10 was suppressed, leading to a significant reduction in the migration of memory TH1 and TH17 lymphocytes across the blood brain barrier (BBB). Our data indicate that laquinimod exerts its therapeutic effects by tightening the BBB and limiting parenchymal invasion of effector T cells, thereby reducing CNS damage.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Neuroprotective Agents/pharmacology , Quinolones/pharmacology , Adult , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/metabolism , Rats, Inbred Lew , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Young Adult
17.
Nat Commun ; 8: 14241, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28117328

ABSTRACT

Multiple Sclerosis (MS) is an inflammatory demyelinating disorder in which remyelination failure contributes to persistent disability. Cholesterol is rate-limiting for myelin biogenesis in the developing CNS; however, whether cholesterol insufficiency contributes to remyelination failure in MS, is unclear. Here, we show the relationship between cholesterol, myelination and neurological parameters in mouse models of demyelination and remyelination. In the cuprizone model, acute disease reduces serum cholesterol levels that can be restored by dietary cholesterol. Concomitant with blood-brain barrier impairment, supplemented cholesterol directly supports oligodendrocyte precursor proliferation and differentiation, and restores the balance of growth factors, creating a permissive environment for repair. This leads to attenuated axon damage, enhanced remyelination and improved motor learning. Remarkably, in experimental autoimmune encephalomyelitis, cholesterol supplementation does not exacerbate disease expression. These findings emphasize the safety of dietary cholesterol in inflammatory diseases and point to a previously unrecognized role of cholesterol in promoting repair after demyelinating episodes.


Subject(s)
Cholesterol, Dietary/administration & dosage , Cholesterol/blood , Multiple Sclerosis/therapy , Myelin Proteins/biosynthesis , Animals , Axons/pathology , Biomarkers/blood , Brain/cytology , Brain/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cholesterol/metabolism , Cholesterol, Dietary/adverse effects , Cuprizone/toxicity , Dietary Supplements , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/blood , Multiple Sclerosis/chemically induced , Oligodendroglia/cytology , Oligodendroglia/pathology , Oligodendroglia/physiology , Primary Cell Culture , Stem Cells/physiology
18.
Proc Natl Acad Sci U S A ; 113(12): 3323-8, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26957602

ABSTRACT

Multiple sclerosis (MS) is caused by T cells that are reactive for brain antigens. In experimental autoimmune encephalomyelitis, the animal model for MS, myelin-reactive T cells initiate the autoimmune process when entering the nervous tissue and become reactivated upon local encounter of their cognate CNS antigen. Thereby, the strength of the T-cellular reactivation process within the CNS tissue is crucial for the manifestation and the severity of the clinical disease. Recently, B cells were found to participate in the pathogenesis of CNS autoimmunity, with several diverse underlying mechanisms being under discussion. We here report that B cells play an important role in promoting the initiation process of CNS autoimmunity. Myelin-specific antibodies produced by autoreactive B cells after activation in the periphery diffused into the CNS together with the first invading pathogenic T cells. The antibodies accumulated in resident antigen-presenting phagocytes and significantly enhanced the activation of the incoming effector T cells. The ensuing strong blood-brain barrier disruption and immune cell recruitment resulted in rapid manifestation of clinical disease. Therefore, myelin oligodendrocyte glycoprotein (MOG)-specific autoantibodies can initiate disease bouts by cooperating with the autoreactive T cells in helping them to recognize their autoantigen and become efficiently reactivated within the immune-deprived nervous tissue.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Central Nervous System Diseases/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Cell Differentiation , Humans , T-Lymphocytes/pathology
19.
Nature ; 530(7590): 349-53, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26863192

ABSTRACT

In multiple sclerosis, brain-reactive T cells invade the central nervous system (CNS) and induce a self-destructive inflammatory process. T-cell infiltrates are not only found within the parenchyma and the meninges, but also in the cerebrospinal fluid (CSF) that bathes the entire CNS tissue. How the T cells reach the CSF, their functionality, and whether they traffic between the CSF and other CNS compartments remains hypothetical. Here we show that effector T cells enter the CSF from the leptomeninges during Lewis rat experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. While moving through the three-dimensional leptomeningeal network of collagen fibres in a random Brownian walk, T cells were flushed from the surface by the flow of the CSF. The detached cells displayed significantly lower activation levels compared to T cells from the leptomeninges and CNS parenchyma. However, they did not represent a specialized non-pathogenic cellular sub-fraction, as their gene expression profile strongly resembled that of tissue-derived T cells and they fully retained their encephalitogenic potential. T-cell detachment from the leptomeninges was counteracted by integrins VLA-4 and LFA-1 binding to their respective ligands produced by resident macrophages. Chemokine signalling via CCR5/CXCR3 and antigenic stimulation of T cells in contact with the leptomeningeal macrophages enforced their adhesiveness. T cells floating in the CSF were able to reattach to the leptomeninges through steps reminiscent of vascular adhesion in CNS blood vessels, and invade the parenchyma. The molecular/cellular conditions for T-cell reattachment were the same as the requirements for detachment from the leptomeningeal milieu. Our data indicate that the leptomeninges represent a checkpoint at which activated T cells are licensed to enter the CNS parenchyma and non-activated T cells are preferentially released into the CSF, from where they can reach areas of antigen availability and tissue damage.


Subject(s)
Cell Movement , Cerebrospinal Fluid/cytology , Encephalomyelitis, Autoimmune, Experimental/pathology , Meninges/pathology , Multiple Sclerosis/pathology , T-Lymphocytes/pathology , Adoptive Transfer , Animals , Cell Adhesion , Cerebrospinal Fluid/immunology , Chemokines/metabolism , Choroid Plexus , Collagen/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Integrin alpha4beta1/metabolism , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Meninges/immunology , Multiple Sclerosis/immunology , Rats , Rats, Inbred Lew , Receptors, CCR5/metabolism , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology
20.
Methods Mol Biol ; 1304: 117-29, 2016.
Article in English | MEDLINE | ID: mdl-25549830

ABSTRACT

The CNS is effectively shielded from the periphery by the blood-brain barrier (BBB) which limits the entry of cells and solutes. However, in autoimmune disorders such as multiple sclerosis, immune cells can overcome this barrier and induce the formation of CNS inflammatory lesions. Recently, two-photon laser scanning microscopy (TPLSM) has made it possible to visualize autoimmune processes in the living CNS in real time. However, along with a high microscopy standard, this technique requires an advanced surgical procedure to access the region of interest. Here, we describe in detail the necessary methodological steps to visualize (auto)immune processes in living rodent tissue. We focus on the procedures to image the leptomeningeal vessels of the thoracic spinal cord during transfer experimental autoimmune encephalomyelitis in LEW rats (AT EAE) and in active EAE in C57BL/6 mice (aEAE).


Subject(s)
Autoimmunity , Central Nervous System/immunology , Animals , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Microscopy, Confocal/methods , Myelin Basic Protein/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Rats , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL