Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
PLoS One ; 9(10): e108756, 2014.
Article in English | MEDLINE | ID: mdl-25286182

ABSTRACT

Inflammatory neuropathies represent disabling human autoimmune disorders with considerable disease variability. Animal models provide insights into defined aspects of their disease pathogenesis. Forkhead box P3 (FoxP3)+ regulatory T lymphocytes (Treg) are anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. Dysfunction or a reduced frequency of Tregs have been associated with different human autoimmune disorders. We here analyzed the functional relevance of Tregs in determining disease manifestation and severity in murine models of autoimmune neuropathies. We took advantage of the DEREG mouse system allowing depletion of Treg with high specificity as well as anti-CD25 directed antibodies to deplete Tregs in mice in actively induced experimental autoimmune neuritis (EAN). Furthermore antibody-depletion was performed in an adoptive transfer model of chronic neuritis. Early Treg depletion increased clinical EAN severity both in active and adoptive transfer chronic neuritis. This was accompanied by increased proliferation of myelin specific T cells and histological signs of peripheral nerve inflammation. Late stage Treg depletion after initial disease manifestation however did not exacerbate inflammatory neuropathy symptoms further. We conclude that Tregs determine disease severity in experimental autoimmune neuropathies during the initial priming phase, but have no major disease modifying function after disease manifestation. Potential future therapeutic approaches targeting Tregs should thus be performed early in inflammatory neuropathies.


Subject(s)
Disease Progression , Forkhead Transcription Factors/metabolism , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies/pharmacology , Disease Models, Animal , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Depletion , Mice, Inbred NOD , Mice, SCID , Myelin Sheath/metabolism , Peripheral Nervous System/pathology
2.
J Biol Chem ; 289(42): 29014-29, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25193661

ABSTRACT

IFNß is a common therapeutic option to treat multiple sclerosis. It is unique among the family of type I IFNs in that it binds to the interferon receptors with high affinity, conferring exceptional biological properties. We have previously reported the generation of an interferon superagonist (dubbed YNSα8) that is built on the backbone of a low affinity IFNα but modified to exhibit higher receptor affinity than even for IFNß. Here, YNSα8 was fused with a 600-residue hydrophilic, unstructured N-terminal polypeptide chain comprising proline, alanine, and serine (PAS) to prolong its plasma half-life via "PASylation." PAS-YNSα8 exhibited a 10-fold increased half-life in both pharmacodynamic and pharmacokinetic assays in a transgenic mouse model harboring the human receptors, notably without any detectable loss in biological potency or bioavailability. This long-lived superagonist conferred significantly improved protection from MOG35-55-induced experimental autoimmune encephalomyelitis compared with IFNß, despite being injected with a 4-fold less frequency and at an overall 16-fold lower dosage. These data were corroborated by FACS measurements showing a decrease of CD11b(+)/CD45(hi) myeloid lineage cells detectable in the CNS, as well as a decrease in IBA(+) cells in spinal cord sections determined by immunohistochemistry for PAS-YNSα8-treated animals. Importantly, PAS-YNSα8 did not induce antibodies upon repeated administration, and its biological efficacy remained unchanged after 21 days of treatment. A striking correlation between increased levels of CD274 (PD-L1) transcripts from spleen-derived CD4(+) cells and improved clinical response to autoimmune encephalomyelitis was observed, indicating that, at least in this mouse model of multiple sclerosis, CD274 may serve as a biomarker to predict the effectiveness of IFN therapy to treat this complex disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon Type I/agonists , Interferon Type I/pharmacology , Peptides/chemistry , Animals , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Flow Cytometry , Humans , Interferon-beta/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/drug therapy , Protein Engineering/methods , Recombinant Proteins/chemistry , Surface Plasmon Resonance , Treatment Outcome
3.
PLoS One ; 9(1): e84259, 2014.
Article in English | MEDLINE | ID: mdl-24416207

ABSTRACT

We have generated transgenic mice that harbor humanized type I interferon receptors (IFNARs) enabling the study of type I human interferons (Hu-IFN-Is) in mice. These "HyBNAR" (Hybrid IFNAR) mice encode transgenic variants of IFNAR1 and IFNAR2 with the human extracellular domains being fused to transmembrane and cytoplasmic segments of mouse sequence. B16F1 mouse melanoma cells harboring the HyBNAR construct specifically bound Hu-IFN-Is and were rendered sensitive to Hu-IFN-I stimulated anti-proliferation, STAT1 activation and activation of a prototypical IFN-I response gene (MX2). HyBNAR mice were crossed with a transgenic strain expressing the luciferase reporter gene under the control of the IFN-responsive MX2 promoter (MX2-Luciferase). Both the HyBNAR and HyBNAR/MX2-Luciferase mice were responsive to all Hu-IFN-Is tested, inclusive of IFNα2A, IFNß, and a human superagonist termed YNSα8. The mice displayed dose-dependent pharmacodynamic responses to Hu-IFN-I injection, as assessed by measuring the expression of IFN-responsive genes. Our studies also demonstrated a weak activation of endogenous mouse interferon response, especially after high dose administration of Hu-IFNs. In sharp contrast to data published for humans, our pharmacodynamic readouts demonstrate a very short-lived IFN-I response in mice, which is not enhanced by sub-cutaneous (SC) injections in comparison to other administration routes. With algometric differences between humans and mice taken into account, the HyBNAR mice provides a convenient non-primate pre-clinical model to advance the study of human IFN-Is.


Subject(s)
Interferon Type I/metabolism , Animals , Cell Line , Homozygote , Humans , Interferon Type I/pharmacology , Luciferases/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/drug effects , Species Specificity , Transfection , Transgenes
4.
Clin Immunol ; 142(3): 351-61, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22264636

ABSTRACT

Parasitic infections frequently lead to immune deviation or suppression. However, the application of specific parasitic molecules in regulating autoimmune responses remains to be explored. Here we report on the immune modulatory function of Lacto-N-fucopentaose III (LNFPIII), a schistosome glycan, in an animal model for multiple sclerosis. We found that LNFPIII treatment significantly reduced the severity of experimental autoimmune encephalomyelitis (EAE) and CNS inflammation, and skewed peripheral immune response to a Th2 dominant profile. Inflammatory monocytes (IMCs) purified from LNFPIII-treated mice had increased expression of nitric oxide synthase 2, and mediated T cell suppression. LNFPIII treatment also significantly increased mRNA expression of arginase-1, aldehyde dehydrogenase 1 subfamily A2, indoleamine 2,3-dioxygenase and heme oxygenase 1 in splenic IMCs. Furthermore, LNFPIII treatment significantly reduced trafficking of dendritic cells across brain endothelium in vitro. In summary, our study demonstrates that LNFPIII glycan treatment suppresses EAE by modulating both innate and T cell immune response.


Subject(s)
Amino Sugars/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Polysaccharides/therapeutic use , Animals , Cell Movement , Dendritic Cells/cytology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunity, Innate , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Nitric Oxide/immunology , Phenotype , T-Lymphocytes/immunology
5.
Eur J Immunol ; 40(12): 3581-90, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21108477

ABSTRACT

Migration of immune cells characterizes inflammation and plays a key role in autoimmune diseases such as MS. CD4(+)Foxp3(+) regulatory T cells (Treg) have the potential to dampen immune responses but show functional impairment in patients with MS. We here show that murine Treg exhibit higher constitutive cell motility in horizontal migration on laminin, surpass non-Treg in transwell assays through microporous membranes as well as across primary brain endothelium and are present in the naïve CNS to a significantly higher extent compared to spleen, lymph nodes and blood. Likewise, human Treg from healthy donors significantly exceed non-Treg in migratory rates across primary human brain endothelium. Finally, we investigated whether the propensity to migrate is impaired as a feature of autoimmunity and therefore tested patients with MS. Treg from patients with stable relapsing-remitting MS show significantly impaired migratory capacity under non-inflammatory conditions compared to healthy donors. We hypothesize that the enhanced propensity to migrate is a feature of Treg that allows for an equilibrium in parenchymal immune surveillance, e.g. of the CNS. Impaired Treg migration across the intact blood-brain barrier, as observed for Treg from patients with MS, indicates a broader functional deficiency hypothetically contributing to early CNS lesion development or phases of MS remissions.


Subject(s)
Blood-Brain Barrier/immunology , Brain/pathology , Cell Movement/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/metabolism , Adult , Animals , Brain/immunology , CD4 Antigens/biosynthesis , Cell Migration Assays, Leukocyte , Cells, Cultured , Female , Forkhead Transcription Factors/biosynthesis , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
6.
J Immunol ; 184(9): 5368-74, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20357264

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are instrumental in peripheral T cell tolerance and innate immunity. How pDCs control peripheral immunetolerance and local parenchymal immune response and contribute to the altered immunoregulation in autoimmune disorders in humans is poorly understood. Based on their surface markers, cytokine production, and ability to prime naive allogenic T cells, we found that purified BDCA-2(+)BDCA-4(+) pDCs consist of at least two separate populations, which differed in their response to oligodeoxynucleotides and IFNs (IFN-beta), and differently induced IL-17- or IL-10-producing T cells. To evaluate the potential immunoregulatory role of these two types of pDCs in multiple sclerosis (MS) and other human autoimmune disorders (myasthenia gravis), we studied the phenotype and regulatory function of pDCs isolated from clinically stable, untreated patients with MS (n = 16). Patients with MS showed a reversed ratio of pDC1/pDC2 in peripheral blood (4.4:1 in healthy controls, 0.69:1 in MS), a phenomenon not observed in the other autoimmune disorders. As a consequence, MS pDCs had an overall propensity to prime IL-17-secreting cells over IL-10-secreting CD4+ T cells. Immunomodulatory therapy with IFN-beta induced an increase of the pDC1 population in vivo (n = 5). Our data offer a plausible explanation for the disturbed immune tolerance in MS patients and provide evidence that immunomodulatory therapy acts at the level of reconstituting homeostasis of pDC, thus reconstituting the disturbed balance.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/pathology , Immunophenotyping , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/pathology , Cells, Cultured , Coculture Techniques , CpG Islands/immunology , Dendritic Cells/classification , Dendritic Cells/drug effects , Humans , Immunophenotyping/methods , Interferon-beta/therapeutic use , Interleukin-17/biosynthesis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Myasthenia Gravis/immunology , Myasthenia Gravis/pathology , Oligodeoxyribonucleotides/pharmacology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
7.
J Immunol Methods ; 356(1-2): 47-59, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20219468

ABSTRACT

Dendritic cells (DCs) are crucial in the initiation of immune responses and are primary targets in vaccination. Here, we describe fluorescent, carbon magnetic nanoparticles (CMNPs) within the 20-80 nm size range that are non-toxic and preferentially endocytosed by DCs. These attributes allow for DC tracing in vitro, ex vivo and in vivo, by both fluorescence and MRI. We show that CMNPs conjugated with an array of proteins are able to induce strong immune responses in mice. The addition of TLR ligand, CpG, to the CMNPs along with protein results in both T cell activation, but also a selective IFNgamma response. The magnetism afforded by the CMNPs facilitates a simple DC enrichment ex vivo by magnetic means from both secondary lymphoid organs, and sites of chronic inflammation. The magnetic and fluorescent properties of the CMNPs allow for visualization, recovery, and potentially the facilitation of directed DC migration. These particles may support more efficient immunization protocols or new diagnostic assays to characterize functionalities of DCs from patients.


Subject(s)
Carbon/chemistry , Dendritic Cells/chemistry , Magnetics , Nanoparticles/chemistry , Animals , Cell Movement , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , T-Lymphocytes/immunology
8.
J Mol Med (Berl) ; 88(6): 535-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20217033

ABSTRACT

Multiple sclerosis (MS) is a chronic immune-mediated, central nervous system (CNS) demyelinating disease. Clinical and histopathological features suggest an inflammatory etiology involving resident CNS innate cells as well as invading adaptive immune cells. Encephalitogenic myelin-reactive T cells have been implicated in the initiation of an inflammatory cascade, eventually resulting in demyelination and axonal damage (the histological hallmarks of MS). Dendritic cells (DC) have recently emerged as key modulators of this immunopathological cascade, as supported by studies in humans and experimental disease models. In one such model, experimental autoimmune encephalomyelitis (EAE), CNS microvessel-associated DC have been shown to be essential for local antigen recognition by myelin-reactive T cells. Moreover, the functional state and compartmental distribution of DC derived from CNS and associated lymphatics seem to be limiting factors in both the induction and effector phases of EAE. Moreover, DC modulate and balance the recruitment of encephalitogenic and regulatory T cells into CNS tissue. This capacity is critically influenced by DC surface expression of co-stimulatory or co-inhibitory molecules. The fact that DC accumulate in the CNS before T cells and can direct T-cell responses suggests that they are key determinants of CNS autoimmune outcomes. Here we provide a comprehensive review of recent advances in our understanding of CNS-derived DC and their relevance to neuroinflammation.


Subject(s)
Autoimmunity/immunology , Central Nervous System/immunology , Dendritic Cells/immunology , Adaptive Immunity/immunology , Animals , Antigens, CD/immunology , B7-H1 Antigen , Central Nervous System/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Immunity, Innate/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , T-Lymphocytes/immunology
9.
J Immunol Methods ; 354(1-2): 80-4, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20138048

ABSTRACT

Cell-type specific expression of the human diphtheria toxin receptor in generally toxin resistant mice represents an innovative approach for the selective depletion of pre-defined cell populations. We demonstrate that in wildtype mice diphtheria toxin--in concentrations otherwise well tolerated--is highly toxic and lethal together with active immunization irrespective of the immunogenic peptide applied. We found increased lung cellularity as only pathological abnormality. Animal models of inflammatory diseases requiring active immunization including experimental autoimmune encephalomyelitis may thus not be applicable in diphtheria receptor transgenic mice pointing to a major limitation of this otherwise technically interesting approach.


Subject(s)
Diphtheria Toxin/toxicity , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunization/adverse effects , Intercellular Signaling Peptides and Proteins/metabolism , Neuritis, Autoimmune, Experimental/immunology , Animals , Diphtheria Toxin/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Freund's Adjuvant/immunology , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lung/drug effects , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/metabolism , Ovalbumin/immunology , Peptide Fragments/immunology , Pertussis Toxin/immunology , Time Factors , Weight Loss/drug effects
10.
Immunol Lett ; 130(1-2): 36-42, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20005257

ABSTRACT

Mycobacterium bovis BCG is still the most widely used vaccine against tuberculosis and CD8(+) T cells play important roles in fighting infection. We investigated how well antigen is processed and presented to CD8(+) T cells using the same well-characterized CD8(+) T cell epitope SIINFEKL expressed in either a cytoplasmic (GFP-OVA) or secreted (85B-OVA) context from BCG. We report that secreted SIINFEKL from 85B-OVA BCG is presented better than cytoplasmic SIINFEKL expressed by GFP-OVA BCG.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/metabolism , Ovalbumin/metabolism , Animals , BCG Vaccine/immunology , Cytoplasm/drug effects , Flow Cytometry , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spleen/cytology
11.
Ann Neurol ; 66(2): 171-83, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19705413

ABSTRACT

OBJECTIVE: We have recently described a novel population of natural regulatory T cells (T(reg)) that are characterized by the expression of HLA-G and may be found at sites of tissue inflammation (HLA-G(pos) T(reg)). Here we studied the role of these cells in multiple sclerosis (MS), a prototypic autoimmune inflammatory disorder of the central nervous system (CNS). METHODS: Sixty-four patients with different types of MS, 9 patients with other neurological diseases, and 20 healthy donors were included in this study. Inflamed brain lesions from 5 additional untreated MS patients were examined. HLA-G(pos) T(reg) were analyzed in the cerebrospinal fluid (CSF) by flow cytometry and in inflammatory demyelinating lesions of MS brain specimens by immunohistochemistry. Functional capacity was accessed and transmigration was determined using an in vitro model of the human blood-brain barrier (BBB). RESULTS: HLA-G(pos) T(reg) were found enriched in the inflamed CSF of MS patients and in inflammatory demyelinating lesions of MS brain specimens. HLA-G(pos) T(reg) showed a strong propensity to transmigrate across BBB, which was vigorously driven by inflammatory chemokines, and associated with a gain of suppressive capacity upon transmigration. CSF-derived HLA-G(pos) T(reg) of MS patients represented a population of activated central memory activated T cells with an upregulated expression of inflammatory chemokine receptors and exhibiting full suppressive capacity. Unlike natural FoxP3-expressing T(reg), HLA-G(pos) T(reg) derived from peripheral blood were functionally unimpaired in MS. INTERPRETATION: In MS, HLA-G(pos) T(reg) may serve to control potentially destructive immune responses directly at the sites of CNS inflammation and to counterbalance inflammation once specifically recruited to the CNS.


Subject(s)
Brain/immunology , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/physiology , Adult , Aged , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/pathology , Brain/physiopathology , Cell Line , Cell Movement , Female , Flow Cytometry , HLA-G Antigens , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR5/metabolism , Receptors, Cytokine/metabolism , Young Adult
12.
Eur J Immunol ; 39(6): 1536-43, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19424967

ABSTRACT

DC in the CNS have emerged as the major rate-limiting factor for immune invasion and subsequent neuroinflammation during EAE. The mechanism of how this is regulated by brain-localized DC remains unknown. Here, we describe the ability of brain-localized DC expressing B7-H1 molecules to recruit CD8(+) T cells to the site of inflammation. Using intracerebral microinjections of B7-homologue 1-deficient DC, we demonstrate a substantial brain infiltration of CD8(+) T cells displaying a regulatory phenotype (CD122(+)) and function, resulting in a decrease of EAE peak clinical values. The recruitment of regulatory-type CD8(+) T cells into the CNS and the role of brain DC expressing B7-homologue 1 molecules in this process open up the possibility of DC-targeted therapeutic manipulation of neuroinflammatory diseases.


Subject(s)
B7-1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Central Nervous System/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Glycoproteins/metabolism , Peptides/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , B7-H1 Antigen , Brain/cytology , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Central Nervous System/pathology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Female , Glycoproteins/administration & dosage , Glycoproteins/immunology , Immune Tolerance/physiology , Interleukin-2 Receptor beta Subunit/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Receptors, CCR6/metabolism , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Vaccination/methods
13.
J Leukoc Biol ; 86(2): 273-81, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19401389

ABSTRACT

CD4(+) T cells constitutively expressing the immune-tolerogenic HLA-G have been described recently as a new type of nT(reg) (HLA-G(pos) T(reg)) in humans. HLA-G(pos) T(reg) accumulate at sites of inflammation and are potent suppressors of T cell proliferation in vitro, suggesting their role in immune regulation. We here characterize the mechanism of how CD4(+) HLA-G(pos) T(reg) influence autologous HLA-G(neg) T(resp) function. Using a suppression system free of APC, we demonstrate a T-T cell interaction, resulting in suppression of HLA-G(neg) T(resp), which is facilitated by TCR engagement on HLA-G(pos) T(reg). Suppression is independent of cell-cell contact and is reversible, as the removal of HLA-G(pos) T(reg) from the established coculture restored the proliferative capability of responder cells. Further, HLA-G(pos) T(reg)-mediated suppression critically depends on the secretion of IL-10 but not TGF-beta.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Immune Tolerance/physiology , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/metabolism , Adult , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion/immunology , Cell Communication/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Down-Regulation/immunology , Female , HLA-G Antigens , Humans , Lymphocyte Activation/immunology , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Young Adult
14.
J Neurosci ; 29(1): 140-52, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19129392

ABSTRACT

Dendritic cells (DCs) appear in higher numbers within the CNS as a consequence of inflammation associated with autoimmune disorders, such as multiple sclerosis, but the contribution of these cells to the outcome of disease is not yet clear. Here, we show that stimulatory or tolerogenic functional states of intracerebral DCs regulate the systemic activation of neuroantigen-specific T cells, the recruitment of these cells into the CNS and the onset and progression of experimental autoimmune encephalomyelitis (EAE). Intracerebral microinjection of stimulatory DCs exacerbated the onset and clinical course of EAE, accompanied with an early T-cell infiltration and a decreased proportion of regulatory FoxP3-expressing cells in the brain. In contrast, the intracerebral microinjection of DCs modified by tumor necrosis factor alpha induced their tolerogenic functional state and delayed or prevented EAE onset. This triggered the generation of interleukin 10 (IL-10)-producing neuroantigen-specific lymphocytes in the periphery and restricted IL-17 production in the CNS. Our findings suggest that DCs are a rate-limiting factor for neuroinflammation.


Subject(s)
Central Nervous System/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Immune System Phenomena/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/cytology , Central Nervous System Stimulants , Dendritic Cells/classification , Dendritic Cells/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Freund's Adjuvant/adverse effects , Glycoproteins/adverse effects , Immune System Phenomena/drug effects , Interferon-alpha/administration & dosage , Interferon-gamma/metabolism , Interleukin-7/metabolism , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/adverse effects , Picrotoxin/administration & dosage , T-Lymphocytes/classification , T-Lymphocytes/immunology , Time Factors
15.
J Immunol ; 181(9): 6201-12, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941210

ABSTRACT

Multiple sclerosis and an animal model resembling multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), are inflammatory demyelinating diseases of the CNS that are suppressed by systemic mycobacterial infection in mice and BCG vaccination in humans. Host defense responses against Mycobacterium in mice are influenced by T lymphocytes and their cytokine products, particularly IFN-gamma, which plays a protective regulatory role in EAE. To analyze the counter-regulatory role of mycobacterial infection-induced IFN-gamma in the CNS on the function of the pathological Th17 cells and the clinical outcome of EAE, we induced EAE in mice that were intracerebrally infected with Mycobacterium bovis bacille Calmette-Guerin (BCG). In this study, we demonstrate that intracerebral (i.c.) BCG infection prevented inflammatory cell recruitment to the spinal cord and suppressed the development of EAE. Concomitantly, there was a significant decrease in the frequency of myelin oligodendrocyte glycoprotein-specific IFN-gamma-producing CD4(+) T cells in the CNS. IL-17(+)CD4(+) T cell responses were significantly suppressed in i.c. BCG-infected mice following EAE induction regardless of T cell specificity. The frequency of Foxp3(+)CD4(+) T cells in these mice was equivalent to that of control mice. Intracerebral BCG infection-induced protection of EAE and suppression of myelin oligodendrocyte glycoprotein-specific IL-17(+)CD4(+) T cell responses were similar in both wild-type and IFN-gamma-deficient mice. These data show that live BCG infection in the brain suppresses CNS autoimmunity. These findings also reveal that the regulation of Th17-mediated autoimmunity in the CNS can be independent of IFN-gamma-mediated mechanisms.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interferon-gamma/physiology , Interleukin-17/antagonists & inhibitors , Mycobacterium bovis/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Tuberculosis/immunology , Animals , Brain/immunology , Brain/metabolism , Brain/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Interferon-gamma/biosynthesis , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-17/biosynthesis , Interleukin-17/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Tuberculosis/pathology
16.
PLoS One ; 3(9): e3149, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18773080

ABSTRACT

In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE), impairment of glial "Excitatory Amino Acid Transporters" (EAATs) together with an excess glutamate-release by invading immune cells causes excitotoxic damage of the central nervous system (CNS). In order to identify pathways to dampen excitotoxic inflammatory CNS damage, we assessed the effects of a beta-lactam antibiotic, ceftriaxone, reported to enhance expression of glial EAAT2, in "Myelin Oligodendrocyte Glycoprotein" (MOG)-induced EAE. Ceftriaxone profoundly ameliorated the clinical course of murine MOG-induced EAE both under preventive and therapeutic regimens. However, ceftriaxone had impact neither on EAAT2 protein expression levels in several brain areas, nor on the radioactive glutamate uptake capacity in a mixed primary glial cell-culture and the glutamate-induced uptake currents in a mammalian cell line mediated by EAAT2. Moreover, the clinical effect of ceftriaxone was preserved in the presence of the EAAT2-specific transport inhibitor, dihydrokainate, while dihydrokainate alone caused an aggravated EAE course. This demonstrates the need for sufficient glial glutamate uptake upon an excitotoxic autoimmune inflammatory challenge of the CNS and a molecular target of ceftriaxone other than the glutamate transporter. Ceftriaxone treatment indirectly hampered T cell proliferation and proinflammatory INFgamma and IL17 secretion through modulation of myelin-antigen presentation by antigen-presenting cells (APCs) e.g. dendritic cells (DCs) and reduced T cell migration into the CNS in vivo. Taken together, we demonstrate, that a beta-lactam antibiotic attenuates disease course and severity in a model of autoimmune CNS inflammation. The mechanisms are reduction of T cell activation by modulation of cellular antigen-presentation and impairment of antigen-specific T cell migration into the CNS rather than or modulation of central glutamate homeostasis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Central Nervous System/drug effects , Inflammation , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , beta-Lactams/pharmacology , Amino Acid Transport System X-AG/metabolism , Animals , Ceftriaxone/pharmacology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Neuroglia/cytology , Rats
17.
Proc Natl Acad Sci U S A ; 105(35): 13015-20, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728195

ABSTRACT

The semaphorin and plexin family of ligand and receptor proteins provides important axon guidance cues required for development. Recent studies have expanded the role of semaphorins and plexins in the regulation of cardiac, circulatory and immune system function. Within the immune system, semaphorins and plexins regulate cell-cell interactions through a complex network of receptor and ligand pairs. Immune cells at different stages of development often express multiple semaphorins and plexins, leading to multivariate interactions, involving more than one ligand and receptor within each functional group. Because of this complexity, the significance of semaphorin and plexin regulation on individual immune cell types has yet to be fully appreciated. In this work, we examined the regulation of T cells by semaphorin 6D. Both in vitro and in vivo T cell stimulation enhanced semaphorin 6D expression. However, semaphorin 6D was only expressed by a majority of T cells during the late phases of activation. Consequently, the targeted disruption of semaphorin 6D receptor-ligand interactions inhibited T cell proliferation at late but not early phases of activation. This proliferation defect was associated with reduced linker of activated T cells protein phosphorylation, which may reflect semaphorin 6D regulation of c-Abl kinase activity. Semaphorin 6D disruption also inhibited expression of CD127, which is required during the multiphase antigen-presenting cell and T cell interactions leading to selection of long-lived lymphocytes. This work reveals a role for semaphorin 6D as a regulator of the late phase of primary immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunity/immunology , Semaphorins/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Semaphorins/antagonists & inhibitors , Signal Transduction
18.
Hum Immunol ; 69(11): 797-804, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18723060

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) of putative autoimmune origin. Recent evidence indicates that MS autoimmunity is linked to defects in regulatory T-cell function, which normally regulates immune responses to self-antigens and prevents autoimmune diseases. MS and its animal model, experimental autoimmune encephalomyelitis (EAE), have long been regarded as a CD4(+) T-cell-mediated autoimmune disease. Studies addressing the role of CD8(+) T cells, however, have only recently begun to emerge. Pathogenic function was attributed to CD8(+) T cells because of their abundant presence or oligoclonal repertoire within MS lesions. However, CD8(+) T cells appeared to have important regulatory functions, as demonstrated in EAE or human MS studies. We here review the contribution of CD8(+) T cells to inflammation and immune regulation in CNS autoimmunity. The knowledge of distinct CD8(+) T-cell populations exerting destructive versus beneficial functions is summarized. The long-term goal is to delineate the exact phenotypic and functional characteristics of regulatory CD8(+) T-cell populations (natural as well as inducible) in humans. This knowledge may help to further develop concepts of reconstituting or enhancing endogenous mechanisms of immune tolerance in future therapeutic concepts for MS.


Subject(s)
Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance , Multiple Sclerosis/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Multiple Sclerosis/pathology , T-Lymphocyte Subsets/pathology
19.
Nat Clin Pract Neurol ; 4(7): 384-98, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18578001

ABSTRACT

The dysregulation of inflammatory responses and of immune self-tolerance is considered to be a key element in the autoreactive immune response in multiple sclerosis (MS). Regulatory T (T(REG)) cells have emerged as crucial players in the pathogenetic scenario of CNS autoimmune inflammation. Targeted deletion of T(REG) cells causes spontaneous autoimmune disease in mice, whereas augmentation of T(REG)-cell function can prevent the development of or alleviate variants of experimental autoimmune encephalomyelitis, the animal model of MS. Recent findings indicate that MS itself is also accompanied by dysfunction or impaired maturation of T(REG) cells. The development and function of T(REG) cells is closely linked to dendritic cells (DCs), which have a central role in the activation and reactivation of encephalitogenic cells in the CNS. DCs and T(REG) cells have an intimate bidirectional relationship, and, in combination with other factors and cell types, certain types of DCs are capable of inducing T(REG) cells. Consequently, T(REG) cells and DCs have been recognized as potential therapeutic targets in MS. This Review compiles the current knowledge on the role and function of various subsets of T(REG) cells in MS and experimental autoimmune encephalomyelitis. We also highlight the role of tolerogenic DCs and their bidirectional interaction with T(REG) cells during CNS autoimmunity.


Subject(s)
Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , T-Lymphocytes, Regulatory/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Multiple Sclerosis/therapy , Self Tolerance/immunology
20.
Eur J Immunol ; 38(6): 1734-44, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18421793

ABSTRACT

The co-inhibitory B7-homologue 1 (B7-H1/PD-L1) influences adaptive immune responses and has been proposed to contribute to the mechanisms maintaining peripheral tolerance and limiting inflammatory damage in parenchymal organs. To understand the B7-H1/PD1 pathway in CNS inflammation, we analyzed adaptive immune responses in myelin oligodendrocyte glycoprotein (MOG)(35-55)-induced EAE and assessed the expression of B7-H1 in human CNS tissue. B7-H1(-/-) mice exhibited an accelerated disease onset and significantly exacerbated EAE severity, although absence of B7-H1 had no influence on MOG antibody production. Peripheral MOG-specific IFN-gamma/IL-17 T cell responses occurred earlier and enhanced in B7-H1(-/-) mice, but ceased more rapidly. In the CNS, however, significantly higher numbers of activated neuroantigen-specific T cells persisted during all stages of EAE. Experiments showing a direct inhibitory role of APC-derived B7-H1 on the activation of MOG-specific effector cells support the assumption that parenchymal B7-H1 is pivotal for delineating T cell fate in the target organ. Compatible with this concept, our data investigating human brain tissue specimens show a strong up-regulation of B7-H1 in lesions of multiple sclerosis. Our findings demonstrate the critical importance of B7-H1 as an immune-inhibitory molecule capable of down-regulating T cell responses thus contributing to the confinement of immunopathological damage.


Subject(s)
B7-1 Antigen/immunology , Central Nervous System/immunology , Membrane Glycoproteins/immunology , Multiple Sclerosis/immunology , Myelin-Associated Glycoprotein/immunology , Peptides/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/analysis , Apoptosis/immunology , B7-H1 Antigen , Cell Count , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Glycoproteins/immunology , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Kinetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...