Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(1): e2209944120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36574650

ABSTRACT

After natalizumab (NAT) cessation, some multiple sclerosis (MS) patients experience a severe disease rebound. The rebound pathophysiology is still unclear; however, it has been linked to interleukin-17-producing T-helper (Th17) cells. We demonstrate that during NAT treatment, MCAM+CCR6+Th17 cells gradually acquire a pathogenic profile, including proinflammatory cytokine production, pathogenic transcriptional signatures, brain endothelial barrier impairment, and oligodendrocyte damage via induction of apoptotic pathways. This is accompanied by an increase in Th17 cell frequencies in the cerebrospinal fluid of NAT-treated patients. Notably, Th17 cells derived from NAT-treated patients, who later developed a disease rebound upon treatment cessation, displayed a distinct transcriptional pathogenicity profile associated with altered migratory properties. Accordingly, increased brain infiltration of patient Th17 cells was illustrated in a humanized mouse model and brain histology from a rebound patient. Therefore, peripheral blood-accumulated MCAM+CCR6+Th17 cells might be involved in rebound pathophysiology, and monitoring of changes in Th17 cell pathogenicity in patients before/during NAT treatment cessation might enable rebound risk assessment in the future.


Subject(s)
Multiple Sclerosis , Th17 Cells , Animals , Mice , Natalizumab/pharmacology , Natalizumab/therapeutic use , Virulence , Multiple Sclerosis/drug therapy , Multiple Sclerosis/cerebrospinal fluid , Brain
2.
Sci Adv ; 6(49)2020 12.
Article in English | MEDLINE | ID: mdl-33277253

ABSTRACT

Remyelination failure in multiple sclerosis (MS) is associated with a migration/differentiation block of oligodendroglia. The reason for this block is highly debated. It could result from disease-related extrinsic or intrinsic regulators in oligodendroglial biology. To avoid confounding immune-mediated extrinsic effect, we used an immune-deficient mouse model to compare induced pluripotent stem cell-derived oligodendroglia from MS and healthy donors following engraftment in the developing CNS. We show that the MS-progeny behaves and differentiates into oligodendrocytes to the same extent as controls. They generate equal amounts of myelin, with bona fide nodes of Ranvier, and promote equal restoration of their host slow conduction. MS-progeny expressed oligodendrocyte- and astrocyte-specific connexins and established functional connections with donor and host glia. Thus, MS oligodendroglia, regardless of major immune manipulators, are intrinsically capable of myelination and making functional axo-glia/glia-glia connections, reinforcing the view that the MS oligodendrocyte differentiation block is not from major intrinsic oligodendroglial deficits.

3.
Acta Neuropathol Commun ; 8(1): 207, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33256847

ABSTRACT

Autoimmune disorders of the central nervous system (CNS) comprise a broad spectrum of clinical entities. The stratification of patients based on the recognized autoantigen is of great importance for therapy optimization and for concepts of pathogenicity, but for most of these patients, the actual target of their autoimmune response is unknown. Here we investigated oligodendrocyte myelin glycoprotein (OMGP) as autoimmune target, because OMGP is expressed specifically in the CNS and there on oligodendrocytes and neurons. Using a stringent cell-based assay, we detected autoantibodies to OMGP in serum of 8/352 patients with multiple sclerosis, 1/28 children with acute disseminated encephalomyelitis and unexpectedly, also in one patient with psychosis, but in none of 114 healthy controls. Since OMGP is GPI-anchored, we validated its recognition also in GPI-anchored form. The autoantibodies to OMGP were largely IgG1 with a contribution of IgG4, indicating cognate T cell help. We found high levels of soluble OMGP in human spinal fluid, presumably due to shedding of the GPI-linked OMGP. Analyzing the pathogenic relevance of autoimmunity to OMGP in an animal model, we found that OMGP-specific T cells induce a novel type of experimental autoimmune encephalomyelitis dominated by meningitis above the cortical convexities. This unusual localization may be directed by intrathecal uptake and presentation of OMGP by meningeal phagocytes. Together, OMGP-directed autoimmunity provides a new element of heterogeneity, helping to improve the stratification of patients for diagnostic and therapeutic purposes.


Subject(s)
Autoantibodies/immunology , Autoimmunity/immunology , Encephalomyelitis, Acute Disseminated/immunology , Multiple Sclerosis/immunology , Oligodendrocyte-Myelin Glycoprotein/immunology , Adult , Animals , Case-Control Studies , Child , Child, Preschool , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Middle Aged , Psychotic Disorders/immunology , Rats , T-Lymphocytes/immunology , Young Adult
4.
Acta Neuropathol ; 140(5): 715-736, 2020 11.
Article in English | MEDLINE | ID: mdl-32894330

ABSTRACT

Multiple sclerosis (MS) is the most frequent demyelinating disease in young adults and despite significant advances in immunotherapy, disease progression still cannot be prevented. Promotion of remyelination, an endogenous repair mechanism resulting in the formation of new myelin sheaths around demyelinated axons, represents a promising new treatment approach. However, remyelination frequently fails in MS lesions, which can in part be attributed to impaired differentiation of oligodendroglial progenitor cells into mature, myelinating oligodendrocytes. The reasons for impaired oligodendroglial differentiation and defective remyelination in MS are currently unknown. To determine whether intrinsic oligodendroglial factors contribute to impaired remyelination in relapsing-remitting MS (RRMS), we compared induced pluripotent stem cell-derived oligodendrocytes (hiOL) from RRMS patients and controls, among them two monozygous twin pairs discordant for MS. We found that hiOL from RRMS patients and controls were virtually indistinguishable with respect to remyelination-associated functions and proteomic composition. However, while analyzing the effect of extrinsic factors we discovered that supernatants of activated peripheral blood mononuclear cells (PBMCs) significantly inhibit oligodendroglial differentiation. In particular, we identified CD4+ T cells as mediators of impaired oligodendroglial differentiation; at least partly due to interferon-gamma secretion. Additionally, we observed that blocked oligodendroglial differentiation induced by PBMC supernatants could not be restored by application of oligodendroglial differentiation promoting drugs, whereas treatment of PBMCs with the immunomodulatory drug teriflunomide prior to supernatant collection partly rescued oligodendroglial differentiation. In summary, these data indicate that the oligodendroglial differentiation block is not due to intrinsic oligodendroglial factors but rather caused by the inflammatory environment in RRMS lesions which underlines the need for drug screening approaches taking the inflammatory environment into account. Combined, these findings may contribute to the development of new remyelination promoting strategies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Oligodendroglia/pathology , Remyelination/immunology , Cell Differentiation/physiology , Humans , Induced Pluripotent Stem Cells , Interferon-gamma/immunology , Oligodendrocyte Precursor Cells/pathology
5.
Acta Neuropathol ; 140(3): 359-375, 2020 09.
Article in English | MEDLINE | ID: mdl-32710244

ABSTRACT

Multiple sclerosis (MS) is the most frequent demyelinating disease and a leading cause for disability in young adults. Despite significant advances in immunotherapies in recent years, disease progression still cannot be prevented. Remyelination, meaning the formation of new myelin sheaths after a demyelinating event, can fail in MS lesions. Impaired differentiation of progenitor cells into myelinating oligodendrocytes may contribute to remyelination failure and, therefore, the development of pharmacological approaches which promote oligodendroglial differentiation and by that remyelination, represents a promising new treatment approach. However, this generally accepted concept has been challenged recently. To further understand mechanisms contributing to remyelination failure in MS, we combined detailed histological analyses assessing oligodendroglial cell numbers, presence of remyelination as well as the inflammatory environment in different MS lesion types in white matter with in vitro experiments using induced-pluripotent stem cell (iPSC)-derived oligodendrocytes (hiOL) and supernatants from polarized human microglia. Our findings suggest that there are multiple reasons for remyelination failure in MS which are dependent on lesion stage. These include lack of myelin sheath formation despite the presence of mature oligodendrocytes in a subset of active lesions as well as oligodendroglial loss and a hostile tissue environment in mixed active/inactive lesions. Therefore, we conclude that better in vivo and in vitro models which mimic the pathological hallmarks of the different MS lesion types are required for the successful development of remyelination promoting drugs.


Subject(s)
Cell Differentiation/physiology , Myelin Sheath/pathology , Oligodendroglia/metabolism , Remyelination/physiology , Adult , Aged , Demyelinating Diseases/pathology , Humans , Microglia/pathology , Middle Aged , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Stem Cells/metabolism
6.
Glia ; 67(5): 870-883, 2019 05.
Article in English | MEDLINE | ID: mdl-30623969

ABSTRACT

In multiple sclerosis, demyelination occurs as a consequence of chronic autoimmunity in the central nervous system causing progressive neurological impairment in patients. After a demyelinating event, new myelin sheaths are formed by adult oligodendroglial progenitor cells; a process called remyelination. However, remyelination often fails in multiple sclerosis due to insufficient recruitment and differentiation of oligodendroglial precursor cells. A pivotal role for the two-pore-domain potassium (K2P ) channel, TASK1, has already been proven for an animal model of multiple sclerosis. However, the mechanisms underlying the TASK1-mediated effects are still elusive. Here, we tested the role of TASK1 channels in oligodendroglial differentiation and remyelination after cuprizone-induced demyelination in male mice. We found TASK1 channels to be functionally expressed on primary murine and human, pluripotent stem cell-derived oligodendrocytes. Lack of TASK1 channels resulted in an increase of mature oligodendrocytes in vitro as well as a higher number of mature oligodendrocytes and accelerated developmental myelination in vivo. Mechanistically, Task1-deficient cells revealed a higher amount of phosphorylated WNK1, a kinase known to be involved in the downstream signaling of the myelination regulator LINGO-1. Furthermore, we analyzed the effect of genetic TASK1 ablation or pharmacological TASK1 inhibition on disease-related remyelination. Neither channel inhibition nor lack of TASK1 channels promoted remyelination after pathological demyelination. In summary, we conclude that functional TASK1 channels participate in the modulation of differentiating oligodendroglial cells in a previously unknown manner. However, while being involved in developmental myelination our data suggest that TASK1 channels have no major effect on remyelination.


Subject(s)
Cell Differentiation/genetics , Demyelinating Diseases/pathology , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Anesthetics, Local/pharmacology , Animals , Animals, Newborn , Bupivacaine/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase Inhibitors/toxicity , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Proteins/ultrastructure , Nerve Tissue Proteins/genetics , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/physiology , Oligodendrocyte Precursor Cells/ultrastructure , Oligodendroglia/drug effects , Oligodendroglia/physiology , Oligodendroglia/ultrastructure , Potassium Channels, Tandem Pore Domain/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Transcription Factors/metabolism , Transcription Factors/pharmacology
8.
Nat Commun ; 9(1): 899, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29500351

ABSTRACT

Oligodendrocytes produce myelin for rapid transmission and saltatory conduction of action potentials in the vertebrate central nervous system. Activation of the myelination program requires several transcription factors including Sox10, Olig2, and Nkx2.2. Functional interactions among them are poorly understood and important components of the regulatory network are still unknown. Here, we identify Nfat proteins as Sox10 targets and regulators of oligodendroglial differentiation in rodents and humans. Overall levels and nuclear fraction increase during differentiation. Inhibition of Nfat activity impedes oligodendrocyte differentiation in vitro and in vivo. On a molecular level, Nfat proteins cooperate with Sox10 to relieve reciprocal repression of Olig2 and Nkx2.2 as precondition for oligodendroglial differentiation and myelination. As Nfat activity depends on calcium-dependent activation of calcineurin signaling, regulatory network and oligodendroglial differentiation become sensitive to calcium signals. NFAT proteins are also detected in human oligodendrocytes, downregulated in active multiple sclerosis lesions and thus likely relevant in demyelinating disease.


Subject(s)
Calcineurin/metabolism , Cell Differentiation , Myelin Sheath/metabolism , NFATC Transcription Factors/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Signal Transduction , Animals , Conserved Sequence , Evolution, Molecular , Gene Expression Regulation , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Mice , Nuclear Proteins , Oligodendrocyte Transcription Factor 2/metabolism , Rats , SOXE Transcription Factors/metabolism , Transcription Factors/metabolism , Zebrafish Proteins
9.
Sci Transl Med ; 9(419)2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29212715

ABSTRACT

Investigations into brain function and disease depend on the precise classification of neural cell types. Cells of the oligodendrocyte lineage differ greatly in their morphology, but accurate identification has thus far only been possible for oligodendrocyte progenitor cells and mature oligodendrocytes in humans. We find that breast carcinoma amplified sequence 1 (BCAS1) expression identifies an oligodendroglial subpopulation in the mouse and human brain. These cells are newly formed, myelinating oligodendrocytes that segregate from oligodendrocyte progenitor cells and mature oligodendrocytes and mark regions of active myelin formation in development and in the adult. We find that BCAS1+ oligodendrocytes are restricted to the fetal and early postnatal human white matter but remain in the cortical gray matter until old age. BCAS1+ oligodendrocytes are reformed after experimental demyelination and found in a proportion of chronic white matter lesions of patients with multiple sclerosis (MS) even in a subset of patients with advanced disease. Our work identifies a means to map ongoing myelin formation in health and disease and presents a potential cellular target for remyelination therapies in MS.


Subject(s)
Multiple Sclerosis/metabolism , Neoplasm Proteins/metabolism , Oligodendroglia/metabolism , Animals , Demyelinating Diseases , Humans , Mice , Multiple Sclerosis/pathology , Myelin Sheath/metabolism
10.
Proc Natl Acad Sci U S A ; 114(11): E2243-E2252, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28246330

ABSTRACT

Rapid and efficient protocols to generate oligodendrocytes (OL) from human induced pluripotent stem cells (iPSC) are currently lacking, but may be a key technology to understand the biology of myelin diseases and to develop treatments for such disorders. Here, we demonstrate that the induction of three transcription factors (SOX10, OLIG2, NKX6.2) in iPSC-derived neural progenitor cells is sufficient to rapidly generate O4+ OL with an efficiency of up to 70% in 28 d and a global gene-expression profile comparable to primary human OL. We further demonstrate that iPSC-derived OL disperse and myelinate the CNS of Mbpshi/shiRag-/- mice during development and after demyelination, are suitable for in vitro myelination assays, disease modeling, and screening of pharmacological compounds potentially promoting oligodendroglial differentiation. Thus, the strategy presented here to generate OL from iPSC may facilitate the studying of human myelin diseases and the development of high-throughput screening platforms for drug discovery.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Transcription Factors/genetics , Animals , Biomarkers , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cell Death/genetics , Cell Lineage/genetics , Cells, Cultured , Cluster Analysis , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Ectopic Gene Expression , Gene Expression Profiling , Humans , Mice , Mutation , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oxidative Stress , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Transcription Factors/metabolism , Transcriptome , tau Proteins/genetics , tau Proteins/metabolism
11.
Toxins (Basel) ; 8(10)2016 10 13.
Article in English | MEDLINE | ID: mdl-27754355

ABSTRACT

Pertussis toxin (PTx), the major virulence factor of the whooping cough-causing bacterial pathogen Bordetella pertussis, permeabilizes the blood-brain barrier (BBB) in vitro and in vivo. Breaking barriers might promote translocation of meningitis-causing bacteria across the BBB, thereby facilitating infection. PTx activates several host cell signaling pathways exploited by the neonatal meningitis-causing Escherichia coli K1-RS218 for invasion and translocation across the BBB. Here, we investigated whether PTx and E. coli K1-RS218 exert similar effects on MAPK p38, NF-κB activation and transcription of downstream targets in human cerebral endothelial TY10 cells using qRT-PCR, Western blotting, and ELISA in combination with specific inhibitors. PTx and E. coli K1-RS218 activate MAPK p38, but only E. coli K1-RS218 activates the NF-κB pathway. mRNA and protein levels of p38 and NF-κB downstream targets including IL-6, IL-8, CxCL-1, CxCL-2 and ICAM-1 were increased. The p38 specific inhibitor SB203590 blocked PTx-enhanced activity, whereas E. coli K1-RS218's effects were inhibited by the NF-κB inhibitor Bay 11-7082. Further, we found that PTx enhances the adherence of human monocytic THP-1 cells to human cerebral endothelial TY10 cells, thereby contributing to enhanced translocation. These modulations of host cell signaling pathways by PTx and meningitis-causing E. coli support their contributions to pathogen and monocytic THP-1 cells translocation across the BBB.


Subject(s)
Endothelial Cells/drug effects , Escherichia coli , Monocytes/drug effects , Pertussis Toxin/toxicity , Cell Adhesion/drug effects , Cell Line , Cerebrum/cytology , Cytokines/genetics , Cytokines/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Meningitis, Escherichia coli , Monocytes/metabolism , Monocytes/physiology , NF-kappa B/metabolism , Phosphorylation/drug effects , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Biol Chem ; 290(41): 24835-43, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26324705

ABSTRACT

Pertussis toxin (PTx), an AB5 toxin and major virulence factor of the whooping cough-causing pathogen Bordetella pertussis, has been shown to affect the blood-brain barrier. Dysfunction of the blood-brain barrier may facilitate penetration of bacterial pathogens into the brain, such as Escherichia coli K1 (RS218). In this study, we investigated the influence of PTx on blood-brain barrier permissiveness to E. coli infection using human brain-derived endothelial HBMEC and TY10 cells as in vitro models. Our results indicate that PTx acts at several key points of host cell intracellular signaling pathways, which are also affected by E. coli K1 RS218 infection. Application of PTx increased the expression of the pathogen binding receptor gp96. Further, we found an activation of STAT3 and of the small GTPase Rac1, which have been described as being essential for bacterial invasion involving host cell actin cytoskeleton rearrangements at the bacterial entry site. In addition, we showed that PTx induces a remarkable relocation of VE-cadherin and ß-catenin from intercellular junctions. The observed changes in host cell signaling molecules were accompanied by differences in intracellular calcium levels, which might act as a second messenger system for PTx. In summary, PTx not only facilitates invasion of E. coli K1 RS218 by activating essential signaling cascades; it also affects intercellular barriers to increase paracellular translocation.


Subject(s)
Bacterial Translocation/drug effects , Brain/cytology , Endothelial Cells/cytology , Escherichia coli/physiology , Microvessels/cytology , Pertussis Toxin/pharmacology , Signal Transduction/drug effects , Antigens, CD/metabolism , Cadherins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Escherichia coli/drug effects , Humans , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Up-Regulation/drug effects , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...