Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(737): eadi0295, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38446903

ABSTRACT

Anti-CD20 therapy to deplete B cells is highly efficacious in preventing new white matter lesions in patients with relapsing-remitting multiple sclerosis (RRMS), but its protective capacity against gray matter injury and axonal damage is unclear. In a passive experimental autoimmune encephalomyelitis (EAE) model whereby TH17 cells promote brain leptomeningeal immune cell aggregates, we found that anti-CD20 treatment effectively spared myelin content and prevented myeloid cell activation, oxidative damage, and mitochondrial stress in the subpial gray matter. Anti-CD20 treatment increased B cell survival factor (BAFF) in the serum, cerebrospinal fluid, and leptomeninges of mice with EAE. Although anti-CD20 prevented gray matter demyelination, axonal loss, and neuronal atrophy, co-treatment with anti-BAFF abrogated these benefits. Consistent with the murine studies, we observed that elevated BAFF concentrations after anti-CD20 treatment in patients with RRMS were associated with better clinical outcomes. Moreover, BAFF promoted survival of human neurons in vitro. Together, our data demonstrate that BAFF exerts beneficial functions in MS and EAE in the context of anti-CD20 treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Relapsing-Remitting , Humans , Animals , Mice , Neuroprotection , Brain , Gray Matter , Antigen Presentation , Atrophy , Encephalomyelitis, Autoimmune, Experimental/drug therapy
2.
Nat Neurosci ; 25(8): 1104-1112, 2022 08.
Article in English | MEDLINE | ID: mdl-35915177

ABSTRACT

To date, most expression quantitative trait loci (eQTL) studies, which investigate how genetic variants contribute to gene expression, have been performed in heterogeneous brain tissues rather than specific cell types. In this study, we performed an eQTL analysis using single-nuclei RNA sequencing from 192 individuals in eight brain cell types derived from the prefrontal cortex, temporal cortex and white matter. We identified 7,607 eGenes, a substantial fraction (46%, 3,537/7,607) of which show cell-type-specific effects, with strongest effects in microglia. Cell-type-level eQTLs affected more constrained genes and had larger effect sizes than tissue-level eQTLs. Integration of brain cell type eQTLs with genome-wide association studies (GWAS) revealed novel relationships between expression and disease risk for neuropsychiatric and neurodegenerative diseases. For most GWAS loci, a single gene co-localized in a single cell type, providing new clues into disease etiology. Our findings demonstrate substantial contrast in genetic regulation of gene expression among brain cell types and reveal potential mechanisms by which disease risk genes influence brain disorders.


Subject(s)
Genome-Wide Association Study , Nervous System Diseases , Brain , Genetic Predisposition to Disease/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics
3.
Int J Mol Sci ; 23(6)2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35328594

ABSTRACT

Successful therapy with anti-CD20 monoclonal antibodies (mAbs) has reinforced the key role of B cells in the immunopathology of multiple sclerosis (MS). This study aimed to determine the effects of a novel class of anti-CD20 mAbs on vascular and extravascular central nervous system (CNS)-infiltrating B cells in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Male hCD20xhIgR3 mice and wild-type C57BL/6 (B6) mice were immunized with human myelin oligodendrocyte glycoprotein (MOG)1-125 to induce EAE. While hCD20xhIgR3 mice were injected intravenously with an anti-human CD20 mAb (5 mg/kg) (rituximab (a type I anti-CD20 mAb) or obinutuzumab (a type II anti-CD20 mAb), B6 mice received the anti-mouse CD20 antibody 18B12. Neither mAb affected clinical disease or serum antibody levels. Obinutuzumab and rituximab had an impact on splenic and CNS-infiltrated B cells with slightly differential depletion efficacy. Additionally, obinutuzumab had beneficial effects on spinal cord myelination. B cell depletion rates in the 18B12/B6 model were comparable with those observed in obinutuzumab-treated hCD20xhIgR3 mice. Our results demonstrate the usefulness of anti-CD20 mAbs for the modulation of B cell-driven peripheral immune response and CNS pathology, with type II antibodies potentially being superior to type I in the depletion of tissue-infiltrating B cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD20 , Central Nervous System , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/drug therapy , Rituximab/pharmacology , Rituximab/therapeutic use
4.
Article in English | MEDLINE | ID: mdl-33653962

ABSTRACT

OBJECTIVE: Therapies targeting B cells have been used in the clinic for multiple sclerosis (MS). In patients with relapsing MS, anti-CD20 therapy often suppresses relapse activity; yet, their effect on disease progression has been disappointing. Most anti-CD20 therapeutic antibodies are type I, but within the unique microenvironment of the brain, type II antibodies may be more beneficial, as type II antibodies exhibit reduced complement-dependent cytotoxicity and they have an increased capacity to induce direct cell death that is independent of the host immune response. METHODS: We compared the effect of type I with type II anti-CD20 therapy in a new rodent model of secondary progressive MS (SPMS), which recapitulates the principal histopathologic features of MS including meningeal B-cell aggregates. Focal MS-like lesions were induced by injecting heat-killed Mycobacterium tuberculosis into the piriform cortex of MOG-immunized mice. Groups of mice were treated with anti-CD20 antibodies (type I [rituxumab, 10 mg/kg] or type II [GA101, 10 mg/kg]) 4 weeks after lesion initiation, and outcomes were evaluated by immunohistochemistry. RESULTS: Anti-CD20 therapy decreased the extent of glial activation, significantly decreased the number of B and T lymphocytes in the lesion, and resulted in disruption of the meningeal aggregates. Moreover, at the given dose, the type II anti-CD20 therapy was more efficacious than the type I and also protected against neuronal death. CONCLUSIONS: These results indicate that anti-CD20 may be an effective therapy for SPMS with B-cell aggregates and that the elimination of CD20+ B cells alone is sufficient to cause disruption of aggregates in the brain.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , B-Lymphocytes/drug effects , Multiple Sclerosis, Chronic Progressive/drug therapy , Rituximab/pharmacology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antigens, CD20 , Astrocytes/drug effects , B-Lymphocytes/immunology , Brain/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microglia/drug effects
5.
Int J Mol Sci ; 21(18)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32962135

ABSTRACT

B cell-depleting therapies have recently proven to be clinically highly successful in the treatment of multiple sclerosis (MS). This study aimed to determine the effects of the novel type II anti-human CD20 (huCD20) monoclonal antibody (mAb) obinutuzumab (OBZ) on spinal cord degeneration in a B cell-dependent mouse model of MS. Double transgenic huCD20xHIGR3 (CD20dbtg) mice, which express human CD20, were immunised with the myelin fusion protein MP4 to induce experimental autoimmune encephalomyelitis (EAE). Both light and electron microscopy were used to assess myelination and axonal pathology in mice treated with OBZ during chronic EAE. Furthermore, the effects of the already established murine anti-CD20 antibody 18B12 were assessed in C57BL/6 wild-type (wt) mice. In both models (18B12/wt and OBZ/CD20dbtg) anti-CD20 treatment significantly diminished the extent of spinal cord pathology. While 18B12 treatment mainly reduced the extent of axonal pathology, a significant decrease in demyelination and increase in remyelination were additionally observed in OBZ-treated mice. Hence, the data suggest that OBZ could have neuroprotective effects on the CNS, setting the drug apart from the currently available type I anti-CD20 antibodies.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antigens, CD20/immunology , Antineoplastic Agents, Immunological/administration & dosage , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis, Chronic Progressive/drug therapy , Spinal Cord/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD20/metabolism , Axons/drug effects , Axons/immunology , Axons/pathology , B-Lymphocytes/pathology , Chronic Disease/drug therapy , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis, Chronic Progressive/immunology , Multiple Sclerosis, Chronic Progressive/pathology , Myelin Basic Protein/immunology , Myelin Proteolipid Protein/immunology , Neurofilament Proteins/blood , Recombinant Fusion Proteins/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord/ultrastructure
6.
PLoS One ; 15(3): e0229850, 2020.
Article in English | MEDLINE | ID: mdl-32155191

ABSTRACT

Reducing Amyloid ß (Aß) in the brain is of fundamental importance for advancing the therapeutics for Alzheimer`s disease. The endogenous metallopeptidase neprilysin (NEP) has been identified as one of the key Aß-degrading enzymes. Delivery of NEP to the brain by utilizing the Brain Shuttle (BS) transport system offers a promising approach for clearing central Aß. We fused the extracellular catalytic domain of NEP to an active or inactive BS module. The two BS-NEP constructs were used to investigate the pharmacokinetic/pharmacodynamics relationships in the blood and the cerebrospinal fluid (CSF) in dose-response and multiple dosing. As previously shown, NEP was highly effective at degrading Aß in blood but not in the CSF compartment after systemic administration. In contrast, the NEP with an active BS module led to a significant CSF exposure of BS-NEP, followed by substantial Aß reduction in CSF and brain parenchyma. Our data show that a BS module against the transferrin receptor facilitates the transport of an Aß degrading enzyme across the blood-brain barriers to efficiently reduce Aß levels in both CSF and brain.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Brain/metabolism , Neprilysin/pharmacology , Recombinant Fusion Proteins/pharmacology , Amyloid beta-Peptides/deficiency , Animals , Blood-Brain Barrier/metabolism , HEK293 Cells , Humans , Neprilysin/cerebrospinal fluid , Neprilysin/pharmacokinetics , Rats , Rats, Wistar , Recombinant Fusion Proteins/cerebrospinal fluid , Recombinant Fusion Proteins/pharmacokinetics
7.
Neuropharmacology ; 120: 38-55, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-26972827

ABSTRACT

Therapeutic antibodies have essentially been banned from the central nervous system, and are so far limited to use mainly in multiple sclerosis. This is primarily due to the fact that antibody penetration across the blood-brain barrier is very limited, with about only 0.1% of circulating antibodies estimated to reach the brain at steady-state concentration. Nonetheless, advances are being made with conventional antibodies, showing that minimal exposure can act centrally to mediate therapeutic effects. Immunotherapy in Alzheimer's disease is a noteworthy example where antibodies against amyloid-ß are able to reduce brain plaque pathology in preclinical models and humans. However, the advances in using antibodies directed at brain targets have also demonstrated impediments of low brain exposure in achieving clinical benefits, spurring increased attention in technologies designed to improve brain exposure of antibodies. Here we review antibodies in clinical trials for central nervous system disorders. Moreover, we describe some of the efforts to improve the therapeutic efficacy of antibodies by enhancing delivery across the blood-brain barrier. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".


Subject(s)
Antibodies/therapeutic use , Central Nervous System Diseases/drug therapy , Immunologic Factors/therapeutic use , Animals , Blood-Brain Barrier/physiology , Central Nervous System Diseases/immunology , Drug Delivery Systems , Humans
8.
Sci Rep ; 5: 14104, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26411801

ABSTRACT

The blood-brain barrier and the blood-cerebrospinal fluid barrier prevent access of biotherapeutics to their targets in the central nervous system and therefore prohibit the effective treatment of neurological disorders. In an attempt to discover novel brain transport vectors in vivo, we injected a T7 phage peptide library and continuously collected blood and cerebrospinal fluid (CSF) using a cisterna magna cannulated conscious rat model. Specific phage clones were highly enriched in the CSF after four rounds of selection. Validation of individual peptide candidates showed CSF enrichments of greater than 1000-fold. The biological activity of peptide-mediated delivery to the brain was confirmed using a BACE1 peptide inhibitor linked to an identified novel transport peptide which led to a 40% reduction of Amyloid-ß in CSF. These results indicate that the peptides identified by the in vivo phage selection approach could be useful transporters for systemically administrated large molecules into the brain with therapeutic benefits.


Subject(s)
Brain/metabolism , Peptides/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Bacteriophage T7/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Cell Surface Display Techniques , Peptide Library , Peptides/chemistry , Peptides/pharmacokinetics , Position-Specific Scoring Matrices , Rats , Reproducibility of Results
9.
Nat Cell Biol ; 17(8): 994-1003, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26214132

ABSTRACT

The use of human pluripotent stem cells for in vitro disease modelling and clinical applications requires protocols that convert these cells into relevant adult cell types. Here, we report the rapid and efficient differentiation of human pluripotent stem cells into vascular endothelial and smooth muscle cells. We found that GSK3 inhibition and BMP4 treatment rapidly committed pluripotent cells to a mesodermal fate and subsequent exposure to VEGF-A or PDGF-BB resulted in the differentiation of either endothelial or vascular smooth muscle cells, respectively. Both protocols produced mature cells with efficiencies exceeding 80% within six days. On purification to 99% via surface markers, endothelial cells maintained their identity, as assessed by marker gene expression, and showed relevant in vitro and in vivo functionality. Global transcriptional and metabolomic analyses confirmed that the cells closely resembled their in vivo counterparts. Our results suggest that these cells could be used to faithfully model human disease.


Subject(s)
Cell Differentiation , Cell Lineage , Endothelial Cells/physiology , Induced Pluripotent Stem Cells/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Animals , Becaplermin , Biomarkers/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/transplantation , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Human Umbilical Vein Endothelial Cells/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/transplantation , Metabolomics/methods , Mice, Inbred NOD , Mice, SCID , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/transplantation , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Phenotype , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Time Factors , Transcription, Genetic , Transfection , Vascular Endothelial Growth Factor A/pharmacology , Wnt Signaling Pathway/drug effects
10.
Neuron ; 81(1): 49-60, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24411731

ABSTRACT

Although biotherapeutics have vast potential for treating brain disorders, their use has been limited due to low exposure across the blood-brain barrier (BBB). We report that by manipulating the binding mode of an antibody fragment to the transferrin receptor (TfR), we have developed a Brain Shuttle module, which can be engineered into a standard therapeutic antibody for successful BBB transcytosis. Brain Shuttle version of an anti-Aß antibody, which uses a monovalent binding mode to the TfR, increases ß-Amyloid target engagement in a mouse model of Alzheimer's disease by 55-fold compared to the parent antibody. We provide in vitro and in vivo evidence that the monovalent binding mode facilitates transcellular transport, whereas a bivalent binding mode leads to lysosome sorting. Enhanced target engagement of the Brain Shuttle module translates into a significant improvement in amyloid reduction. These findings have major implications for the development of biologics-based treatment of brain disorders.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Protein Transport/physiology , Single-Chain Antibodies/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Amyloid beta-Protein Precursor/genetics , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/immunology , Cell Line, Transformed , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Macrolides/pharmacology , Mice , Mice, Transgenic , Models, Immunological , Presenilin-1/genetics , Protein Binding/drug effects , Protein Binding/immunology , Protein Transport/drug effects , Receptors, Transferrin/immunology , Receptors, Transferrin/metabolism , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/therapeutic use , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors , Transcytosis/drug effects , Transcytosis/genetics , Transcytosis/immunology
11.
Sci Rep ; 3: 1500, 2013.
Article in English | MEDLINE | ID: mdl-23511305

ABSTRACT

The blood brain barrier (BBB) has evolved unique characteristics such as dense coverage of the endothelial cells by pericytes and interactions with astrocytes through perivascular endfeet. We study BBB formation in a 3-dimensional multicellular spheroid system of human primary brain endothelial cells (hpBECs), primary pericytes (hpPs) and primary astrocytes (hpAs). We show for the first time that hpBECs, hpPs and hpAs spontaneously self-organize into a defined multicellular structure which recapitulates the complex arrangement of the individual cell types in the BBB structure. Pericytes play a crucial role mediating the interaction between hpBECs and hpAs. This process is not dependent on a scaffold support demonstrating that formation and cellular architecture of the BBB is intrinsically programmed within each specific cell type. In a matrigel setup the hpBECs, hpPs and hpAs also undergo self-arrangement to form endothelial tube-like structures tightly covered by hpPs and loosely attached hpAs mainly at the junctions.


Subject(s)
Blood-Brain Barrier/cytology , Models, Biological , Spheroids, Cellular/cytology , Astrocytes/cytology , Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Microvessels/cytology , Neovascularization, Physiologic , Pericytes/cytology , Pericytes/metabolism , Receptors, Cell Surface/metabolism , Spheroids, Cellular/metabolism
12.
PLoS One ; 7(5): e38149, 2012.
Article in English | MEDLINE | ID: mdl-22675443

ABSTRACT

Brain microvascular endothelial cells (BEC) constitute the blood-brain barrier (BBB) which forms a dynamic interface between the blood and the central nervous system (CNS). This highly specialized interface restricts paracellular diffusion of fluids and solutes including chemicals, toxins and drugs from entering the brain. In this study we compared the transcriptome profiles of the human immortalized brain endothelial cell line hCMEC/D3 and human primary BEC. We identified transcriptional differences in immune response genes which are directly related to the immortalization procedure of the hCMEC/D3 cells. Interestingly, astrocytic co-culturing reduced cell adhesion and migration molecules in both BECs, which possibly could be related to regulation of immune surveillance of the CNS controlled by astrocytic cells within the neurovascular unit. By matching the transcriptome data from these two cell lines with published transcriptional data from freshly isolated mouse BECs, we discovered striking differences that could explain some of the limitations of using cultured BECs to study BBB properties. Key protein classes such as tight junction proteins, transporters and cell surface receptors show differing expression profiles. For example, the claudin-5, occludin and JAM2 expression is dramatically reduced in the two human BEC lines, which likely explains their low transcellular electric resistance and paracellular leakiness. In addition, the human BEC lines express low levels of unique brain endothelial transporters such as Glut1 and Pgp. Cell surface receptors such as LRP1, RAGE and the insulin receptor that are involved in receptor-mediated transport are also expressed at very low levels. Taken together, these data illustrate that BECs lose their unique protein expression pattern outside of their native environment and display a more generic endothelial cell phenotype. A collection of key genes that seems to be highly regulated by the local surroundings of BEC within the neurovascular unit are presented and discussed.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Transcriptome , Animals , Astrocytes/metabolism , Biological Transport/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Coculture Techniques , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunophenotyping , Interferons/metabolism , Mice , Models, Animal , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Tight Junctions/metabolism
13.
Nat Immunol ; 11(12): 1110-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21037578

ABSTRACT

B lymphocytes differentiate into antibody-secreting cells under the antigen-specific control of follicular helper T cells (T(FH) cells). Here we demonstrate that isotype-switched plasma cells expressed major histocompatibility complex (MHC) class II, the costimulatory molecules CD80 and CD86, and the intracellular machinery required for antigen presentation. Antigen-specific plasma cells accessed, processed and presented sufficient antigen in vivo to induce multiple helper T cell functions. Notably, antigen-primed plasma cells failed to induce interleukin 21 (IL-21) or the transcriptional repressor Bcl-6 in naive helper T cells and actively decreased these key molecules in antigen-activated T(FH) cells. Mice lacking plasma cells showed altered T(FH) cell activity, which provided evidence of this negative feedback loop. Hence, antigen presentation by plasma cells defines a previously unknown layer of cognate regulation that limits the antigen-specific T(FH) cell program that controls ongoing B cell immunity.


Subject(s)
Antigen Presentation/immunology , Lymphocyte Activation/immunology , Plasma Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity , Animals , Cell Separation , Enzyme-Linked Immunospot Assay , Flow Cytometry , Immunologic Memory , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction
14.
Infect Immun ; 75(8): 3842-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17517881

ABSTRACT

Neuroborreliosis (NB) is a chronic infectious disease of the central nervous system (CNS) caused by a tick-borne spirochete, Borrelia burgdorferi. In addition to direct effects of the causative infectious agent, additional immunity-mediated mechanisms are thought to play a role in the CNS pathology of NB. In order to further understand the involvement of humoral immune mechanisms in NB, we dissected the intrathecal antibody responses down to the single-plasma-cell level. Starting with single-cell reverse transcription-PCR of fluorescence-activated cell sorter-sorted cerebrospinal fluid plasma cells from an NB patient, we identified expanded clones and resurrected the antigen specificity of their secreted antibodies through recombinant expression of the correctly paired immunoglobulin heavy- and light-chain genes as monoclonal antibodies (MAbs). As expected, we found specificity for the causative infectious agent, B. burgdorferi, among the clonally expanded plasma cell (cePC)-derived MAbs. However, from an independent cePC of the same patient, we could derive MAbs specific for human CNS myelin, without detectable cross-reactivity with B. burgdorferi antigens. While reactivity against B. burgdorferi is a known feature of humoral immune responses in NB, we show (i) that immune responses specific for self antigens may be a distinct feature of CNS infections independent of pathogen reactivity and (ii) that humoral autoimmunity in NB (since found in cePC) is the result of a truly antigen-driven immune response. Our findings indicate that in NB mechanisms may be at play that induce distinct immune responses specific for pathogen and self antigens independent from "molecular mimicry."


Subject(s)
Autoimmunity , Borrelia burgdorferi/immunology , Borrelia burgdorferi/pathogenicity , Lyme Neuroborreliosis/microbiology , Adult , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigens, Bacterial/immunology , Autoantibodies/immunology , Autoantigens/immunology , Cerebrospinal Fluid/immunology , Cross Reactions , Humans , Lyme Neuroborreliosis/immunology , Male , Microscopy, Fluorescence , Myelin Sheath/immunology , Plasma Cells/immunology
15.
Proc Natl Acad Sci U S A ; 103(49): 18697-702, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17121989

ABSTRACT

The precise mechanisms leading to CNS inflammation and myelin destruction in both multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) remain the subject of intense debate. In both MS and EAE, autoantibodies (autoAbs) are thought to be involved in tissue destruction through recruiting Fc receptor (FcR)-bearing cells or direct cytotoxic effects through the activation of the complement pathway. Whereas intrathecal immunoglobulin (Ig) production and Ig deposition in inflammatory lesions is a hallmark of MS, mice deficient in B cells and Igs develop severe EAE. Paradoxically, mice of the same genetic background but deficient in FcRgamma are EAE-resistant. We found that the functional expression of FcRgamma on systemic accessory cells, but not CNS-resident cells, appears to be vital for the development of CNS inflammation, independent of antigen-presenting cell function or Ab involvement. On the other hand, we found that the injection of antimyelin oligodendrocyte glycoprotein-Abs drastically worsens disease severity, inflammation, and demyelination. Using FcRgamma(-/-) and C1q(-/-) mice, we could definitively establish that the demyelinating capacity of such autoAb in vivo relies entirely on complement activation and is FcR-independent.


Subject(s)
Autoantibodies/physiology , Complement Activation/immunology , Complement System Proteins/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Receptors, IgG/physiology , Animals , Complement Activation/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, IgG/deficiency , Receptors, IgG/genetics
16.
Nat Immunol ; 7(9): 946-53, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16906165

ABSTRACT

T helper type 1 (T(H)1) lymphocytes are considered to be the main pathogenic cell type responsible for organ-specific autoimmune inflammation. As interleukin 18 (IL-18) is a cofactor with IL-12 in promoting T(H)1 cell development, we examined the function of IL-18 and its receptor, IL-18R, in autoimmune central nervous system inflammation. Similar to IL-12-deficient mice, IL-18-deficient mice were susceptible to experimental autoimmune encephalomyelitis. In contrast, IL-18R alpha-deficient mice were resistant to experimental autoimmune encephalomyelitis, indicating involvement of an IL-18R alpha ligand other than IL-18 with encephalitogenic properties. Moreover, engagement of IL-18R alpha on antigen-presenting cells was required for the generation of pathogenic IL-17-producing T helper cells. Thus, IL-18 and T(H)1 cells are dispensable, whereas IL-18R alpha and IL-17-producing T helper cells are required, for autoimmune central nervous system inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Interleukin-17/metabolism , Receptors, Interleukin-18/physiology , Th1 Cells/immunology , Animals , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/immunology , Antigens/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-12/metabolism , Interleukin-18/genetics , Interleukin-18/physiology , Interleukin-23/metabolism , Mice , Mice, Knockout , Mitogens/immunology , Receptors, Interleukin-18/analysis , Receptors, Interleukin-18/genetics
17.
Proc Natl Acad Sci U S A ; 101 Suppl 2: 14670-6, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15292505

ABSTRACT

Prion diseases are characterized by the deposition of an abnormal form (termed PrP(Sc)) of the cellular prion protein (PrP(C)). Because antibodies to PrP(C) can antagonize deposition of PrP(Sc) in cultured cells and mice, they may be useful for anti-prion therapy. However, induction of protective anti-prion immune responses in WT animals may be hindered by host tolerance. Here, we studied the cellular and molecular basis of tolerance to PrP(C). Immunization of Prnp(o/o) mice with bacterially expressed PrP (PrP(REC)) resulted in vigorous humoral immune responses to PrP(REC) and native cell-surface PrP(C). Instead, WT mice yielded antibodies that failed to recognize native PrP(C) despite immunoreactivity with PrP(REC), even after immunization with PrP-PrP polyprotein and/or upon administration of anti-OX40 antibodies. Consequently, immunized WT mice experienced insignificantly delayed prion pathogenesis upon peripheral prion challenge. Anti-PrP immune responses in Prnp(o/o) mice were completely abrogated by transgenic expression of PrP(C) in B cells, T cells, neurons, or hepatocytes, but only moderately reduced by expression in myelinating cells, despite additional thymic Prnp transcription in each case. We conclude that tolerance to PrP(C) can coexist with immunoreactivity to PrP(REC) and does not depend on thymic PrP(C) expression. Its circumvention might represent an important step toward the development of effective anti-prion immunotherapy.


Subject(s)
Prion Diseases/immunology , Prion Diseases/prevention & control , Prions/immunology , Animals , Antibody Formation , Female , Immune Tolerance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , PrPC Proteins/immunology , PrPSc Proteins/immunology , Prion Diseases/therapy , Prions/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...