Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 114(4): 734-739, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28057865

ABSTRACT

Autoantibodies against myelin oligodendrocyte glycoprotein (MOG) are associated with autoimmune central nervous system diseases like acute disseminated encephalomyelitis (ADEM). For ADEM, it is speculated that a preceding infection is the trigger of the autoimmune response, but the mechanism connecting the infection to the production of MOG antibodies remains a mystery. We reasoned that the ability of B cells to capture cognate antigen from cell membranes, along with small quantities of coexpressed "bystander" antigens, might enable B-cell escape from tolerance. We tested this hypothesis using influenza hemagglutinin as a model viral antigen and transgenic, MOG-specific B cells. Using flow cytometry and live and fixed cell microscopy, we show that MOG-specific B cells take up large amounts of MOG from cell membranes. Uptake of the antigen from the membrane leads to a strong activation of the capturing B cell. When influenza hemagglutinin is also present in the membrane of the target cell, it can be cocaptured with MOG by MOG-specific B cells via the B-cell receptor. Hemagglutinin and MOG are both presented to T cells, which in turn are activated and proliferate. As a consequence, MOG-specific B cells get help from hemagglutinin-specific T cells to produce anti-MOG antibodies. In vivo, the transfer of MOG-specific B cells into recipient mice after the cocapture of MOG and hemagglutinin leads to the production of class-switched anti-MOG antibodies, dependent on the presence of hemagglutinin-specific T cells. This mechanism offers a link between infection and autoimmunity.


Subject(s)
Antigens, Viral/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , Animals , Autoantibodies/immunology , Autoimmunity/immunology , Cell Line , Cell Membrane/immunology , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology
2.
J Autoimmun ; 86: 39-50, 2018 01.
Article in English | MEDLINE | ID: mdl-28958667

ABSTRACT

INTRODUCTION: The mode of action of dimethyl fumarate (DMF), an immunomodulatory treatment for relapsing-remitting multiple sclerosis (RRMS), has not yet been fully elucidated. While in-vitro experiments and animal studies suggest effects on immune cell survival, proliferation, migration and oxidative stress response, corresponding observations from human studies are lacking. This study aims to characterize ex-vivo and in-vivo effects in a cohort of DMF treated RRMS patients. METHODS: Blood samples were collected from twenty well-characterized RRMS patients at baseline and after 3, 6 and 12 months of DMF treatment and an age- and gender-matched cohort of 20 healthy individuals at 0 and 3 months. Leukocyte subpopulations, immunoglobulin levels and cytokine secretion were measured. T cells were assessed for their levels of reactive oxygen species (ROS), metabolic status and their proliferative capacity. Levels of antioxidants were determined in serum by mass spectrometry. Responses of monocyte activation markers as well as NFkB and MAPK pathways to DMF were analysed. RESULTS: Upon DMF treatment, all lymphocyte subpopulations dropped significantly over the course of 12 months with cytotoxic and effector T cells being affected most significantly. DMF induced cell death and inhibited proliferation of T cells in-vitro. Interestingly, this anti-proliferative effect decreased under treatment. In-vivo DMF treatment led to decreased T cell glycolysis and higher turn-over of antioxidants. In line with these results a significant increase of cytosolic ROS levels after 3 months treatment was detected in T cells. In-vitro DMF treatment reduced NFkB (p65) translocation to the nucleus and MAPK (p38) levels decreased upon stimulation with monomethyl fumarate (MMF) in-vitro and ex-vivo. Consequently, the expression of co-stimulatory molecules like CD40 and CD150 was decreased in antigen presenting cells both in-vitro and ex-vivo. CONCLUSION: This study translates knowledge from in-vitro and animal studies on DMF into the clinical setting. Our data suggest that DMF not only alters lymphocyte composition, but also has profound effects on proliferation and induces oxidative stress in T cells. It also acts on innate immunity by reducing the activation status of antigen presenting cells (APCs) via NFkB and MAPK inactivation.


Subject(s)
Antigen-Presenting Cells/immunology , Dimethyl Fumarate/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adaptive Immunity , Adult , Antigen Presentation , Antigen-Presenting Cells/drug effects , Cell Death , Cell Proliferation , Cells, Cultured , Cohort Studies , Female , Glycolysis , Humans , Immunity, Innate , Male , Middle Aged , NF-kappa B/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Helper-Inducer/drug effects
3.
J Neuroinflammation ; 12: 46, 2015 Mar 08.
Article in English | MEDLINE | ID: mdl-25889963

ABSTRACT

BACKGROUND: Antibodies against myelin oligodendrocyte glycoprotein (MOG) have been identified in a subgroup of pediatric patients with inflammatory demyelinating disease of the central nervous system (CNS) and in some patients with neuromyelitis optica spectrum disorder (NMOSD). The aim of this study was to examine the frequency, clinical features, and long-term disease course of patients with anti-MOG antibodies in a European cohort of NMO/NMOSD. FINDINGS: Sera from 48 patients with NMO/NMOSD and 48 patients with relapsing-remitting multiple sclerosis (RR-MS) were tested for anti-aquaporin-4 (AQP4) and anti-MOG antibodies with a cell-based assay. Anti-MOG antibodies were found in 4/17 patients with AQP4-seronegative NMO/NMOSD, but in none of the AQP4-seropositive NMO/NMOSD (n = 31) or RR-MS patients (n = 48). MOG-seropositive patients tended towards younger disease onset with a higher percentage of patients with pediatric (<18 years) disease onset (MOG+, AQP4+, MOG-/AQP4-: 2/4, 3/31, 0/13). MOG-seropositive patients presented more often with positive oligoclonal bands (OCBs) (3/3, 5/29, 1/13) and brain magnetic resonance imaging (MRI) lesions during disease course (2/4, 5/31, 1/13). Notably, the mean time to the second attack affecting a different CNS region was longer in the anti-MOG antibody-positive group (11.3, 3.2, 3.4 years). CONCLUSIONS: MOG-seropositive patients show a diverse clinical phenotype with clinical features resembling both NMO (attacks mainly confined to the spinal cord and optic nerves) and MS with an opticospinal presentation (positive OCBs, brain lesions). Anti-MOG antibodies can serve as a diagnostic and maybe prognostic tool in patients with an AQP4-seronegative NMO phenotype and should be tested in those patients.


Subject(s)
Autoantibodies/blood , Myelin-Oligodendrocyte Glycoprotein/immunology , Neuromyelitis Optica/blood , Adult , Aged , Aquaporin 4/immunology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/blood , Retrospective Studies , Young Adult
4.
Mult Scler ; 20(10): 1363-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24598267

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) have emerged as a family of post-transcriptional regulators of gene expression that mediate diverse aspects of immunity. MiRNA dysregulation has been found in multiple sclerosis (MS), reflecting the growing need to identify disease-specific miRNA expression signatures. Our previous low-density array studies reveal differential miR-126 expression in the CD4(+)T cells of untreated relapsing-remitting MS (RRMS) patients. Here, we investigated miR-126 expression in natalizumab-treated patients. METHODS: We isolated CD4(+) T cells from untreated (n = 12) and natalizumab-treated MS patients (n = 24), and from healthy volunteers (n = 12). We analyzed the expression of miRNAs and potential targets by real time reverse transcription polymerase chain reaction (RT-PCR). We assessed specific inhibition of miR-126, in vitro. RESULTS: MiR-126 was down-regulated in cells of patients under natalizumab treatment and up-regulated during relapse, supporting a regulatory role in MS immunopathogenesis. MiR-126 expression correlated with the expression of POU2AF1, a regulator of Spi-B that binds to the promoter/enhancer sequences of JC virus (JCV), the pathogen of progressive multifocal leukoencephalopathy (PML), a rare complication of natalizumab treatment. The same trend was found for Spi-B. Strong up-regulation of both genes appeared to be treatment duration-dependent. Specific inhibition experiments supported the link between the expression of miR-126 and POU2AF1/Spi-B. CONCLUSIONS: Our findings provided deeper insight into the mode of action of natalizumab, with possible implications for understanding both the effects of natalizumab on MS activity and its specific adverse event profile.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , Immunosuppressive Agents/therapeutic use , MicroRNAs/metabolism , Multiple Sclerosis/drug therapy , Adult , Antibodies, Monoclonal, Humanized/adverse effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation , Humans , Immunosuppressive Agents/adverse effects , Male , MicroRNAs/genetics , Middle Aged , Multiple Sclerosis/diagnosis , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Natalizumab , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Treatment Outcome
5.
Clin Immunol ; 144(1): 70-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22659298

ABSTRACT

MicroRNAs (miRNAs) are posttranscriptional regulators of gene expression. We compared the expression of 1059 miRNAs in B lymphocytes from untreated and natalizumab treated relapsing-remitting multiple sclerosis (RRMS) patients and healthy volunteers (HV). Forty nine miRNAs were down-regulated in untreated MS patients compared with HV. A distinct pattern of 10 differentially expressed miRNAs was found in natalizumab treated patients compared with untreated patients. Two clusters, i.e. miR-106b-25 and miR-17-92, were particularly deregulated. MiRNA-mRNA interaction analysis revealed B cell receptor, phosphatidyl-inositol-3-kinase (PI3K) and phosphatase and tensin homology (PTEN) signaling being the key affected pathways. We discovered deregulated viral miRNAs in untreated patients as compared with HV and natalizumab treated patients, a novel finding that may be related to latency and activation of viruses in MS. Our findings provide first insights into miRNA dependent regulation of B cell function in MS and the impact of a therapy not primarily targeting B cells on this regulation.


Subject(s)
B-Lymphocytes/immunology , MicroRNAs/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Female , Humans , Male , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/genetics , Natalizumab
6.
Ann Neurol ; 69(2): 408-13, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21387383

ABSTRACT

T cells exit secondary lymphoid organs along a sphingosine1-phosphate (S1P) gradient and, accordingly, are reduced in blood upon fingolimod-mediated S1P-receptor (S1PR)-blockade. Serving as a model of adaptive immunity, we characterized cellular and humoral immune responses to influenza vaccine in fingolimod-treated patients with multiple sclerosis (MS) and in untreated healthy controls. Although the mode of action of fingolimod might predict reduced immunity, vaccine-triggered T cells accumulated normally in blood despite efficient S1PR-blockade. Concentrations of anti-influenza A/B immunoglobulin (Ig)M and IgG also increased similarly in both groups. These results indicate that fingolimod-treated individuals can mount vaccine-specific adaptive immune responses comparable to healthy controls.


Subject(s)
Adaptive Immunity/immunology , Immunosuppressive Agents/therapeutic use , Influenza Vaccines/immunology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Adult , Enzyme-Linked Immunospot Assay , Female , Fingolimod Hydrochloride , Flow Cytometry , Humans , Male , Middle Aged , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/therapeutic use , T-Lymphocytes/immunology
7.
Epilepsia ; 53(5): 922-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22509781

ABSTRACT

PURPOSE: Status epilepticus (SE) has deleterious effects on brain tissue, but whether brief recurrent seizures may also damage neurons represents a matter of controversy. Therefore, it remains a central area of epilepsy research to identify individuals at risk where disease progression can be potentially prevented. Biomarkers may serve as tools for such identification. Thus the present study aimed at analyzing the levels of heat shock protein 70 (HSP-70, also designated as HSPA1A) and neurofilament heavy chain protein (NfH(SMI35) ) in cerebrospinal fluid (CSF) of patients with seizures of different severity. METHODS: Forty-one patients were included, of whom 20 patients had a single generalized tonic-clonic seizure (GTCS) episode (SS), 11 had repetitive GTCS (RS), and 10 experienced convulsive SE. The control group consisted of 18 subjects. HSP-70 levels were measured using a conventional enzyme-linked immunosorbent assay (ELISA), whereas the NfH(SMI35) protein levels were detected by an electrochemiluminescence (ECL) immunoassay. KEY FINDINGS: Patients with SE (p < 0.001) and RS (p < 0.05) had significantly higher NfH(SMI35) levels than controls, and SE was associated with increased concentrations when compared with SS (p < 0.001). NfH(SMI35) levels in SS did not differ from controls. Patients with SE had significantly raised HSP-70 levels compared to RS (p < 0.05), SS (p < 0.05), and controls (p < 0.001). SS and RS did not differ from each or from controls. Levels of NfH(SMI35) and HSP-70 showed a significant correlation (r = 0.34; p = 0.007) in the group of all study subjects, which was not apparent when controls and patients with seizures were considered separately. The correlation between NfH(SMI35) and HSP-70 tended to be inverse in patients with SE, but it did not reach statistical significance (r = -0.3; p > 0.05). SIGNIFICANCE: Studying biochemical markers as additional quantitative tools for the measurement of neuronal damage (especially subclinical), complementary to available techniques of imaging, and clinical assessment might prove useful for identifying patients at risk of accumulating neuronal injury resulting from uncontrolled seizures. NfH(SMI35) and HSP-70 are of potential value as sensitive and specific biomarkers of seizure-related pathologic events. Future longitudinal studies are needed to monitor such patients by correlating biochemical, neuroimaging, and clinical methods of assessment.


Subject(s)
Brain Injuries/cerebrospinal fluid , Brain Injuries/etiology , HSP70 Heat-Shock Proteins/cerebrospinal fluid , Neurofilament Proteins/cerebrospinal fluid , Seizures/complications , Adult , Aged , Analysis of Variance , Anticonvulsants/therapeutic use , Electroencephalography , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Seizures/drug therapy , Statistics, Nonparametric , Tomography, X-Ray Computed , Young Adult
8.
Hum Mol Genet ; 18(4): 767-78, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19010793

ABSTRACT

Multiple sclerosis (MS), a chronic disorder of the central nervous system and common cause of neurological disability in young adults, is characterized by moderate but complex risk heritability. Here we report the results of a genome-wide association study performed in a 1000 prospective case series of well-characterized individuals with MS and group-matched controls using the Sentrix HumanHap550 BeadChip platform from Illumina. After stringent quality control data filtering, we compared allele frequencies for 551 642 SNPs in 978 cases and 883 controls and assessed genotypic influences on susceptibility, age of onset, disease severity, as well as brain lesion load and normalized brain volume from magnetic resonance imaging exams. A multi-analytical strategy identified 242 susceptibility SNPs exceeding established thresholds of significance, including 65 within the MHC locus in chromosome 6p21.3. Independent replication confirms a role for GPC5, a heparan sulfate proteoglycan, in disease risk. Gene ontology-based analysis shows a functional dichotomy between genes involved in the susceptibility pathway and those affecting the clinical phenotype.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Glypicans/genetics , Multiple Sclerosis/genetics , Adolescent , Adult , Age of Onset , Aged , Case-Control Studies , Child , Female , Humans , Male , Middle Aged , Phenotype , Polymorphism, Single Nucleotide , Prospective Studies , White People/genetics , Young Adult
9.
Eur J Immunol ; 40(3): 888-98, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20148420

ABSTRACT

MicroRNA (miRNA) are a class of post-transcriptional regulators of gene expression targeting mRNA for translational repression and/or degradation. We analyzed the expression of 365 miRNA in lymphocytes in relapsing-remitting MS patients, and show the first evidence for distinct miRNA expression profiles in CD4(+), CD8(+) and B cells in MS when compared with those in healthy volunteers. MiR-17-5p, which is involved in autoimmunity, was up-regulated in CD4(+) cells from MS patients. This was correlated with alterations in the expression of potential target genes of miR-17-5p, i.e. phosphatase and tensin homology and phosphatidyl-inositol-3-kinase regulatory subunit 1, which were down-regulated upon stimulation of CD4(+) cells with anti-CD3/CD28 in vitro. Functional experiments with a synthetic inhibitor of miR-17 supported the link between miRNA expression and the altered target gene expression. Moreover, we found distinct responses of deregulated miRNA to stimulation, i.e. miR-17-5p and miR-193a were strongly up-regulated, in contrast to the down-regulation of miR-497, miR-1 and miR-126. Other deregulated miRNA did not respond to the stimulation probably due to other, non-T-cell activation related, mechanisms in their mode of action. Our findings support the role of miRNA-dependent regulatory mechanisms in the immunopathogenesis of MS.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Gene Expression Profiling , MicroRNAs/biosynthesis , Multiple Sclerosis, Relapsing-Remitting/genetics , B-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Separation , Female , Flow Cytometry , Gene Expression , Humans , Male , MicroRNAs/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Front Immunol ; 10: 829, 2019.
Article in English | MEDLINE | ID: mdl-31040853

ABSTRACT

Determining antigen specificity is vital for understanding B cell biology and for producing human monoclonal antibodies. We describe here a powerful method for identifying B cells that recognize membrane antigens expressed on cells. The technique depends on two characteristics of the interaction between a B cell and an antigen-expressing cell: antigen-receptor-mediated extraction of antigen from the membrane of the target cell, and B cell activation. We developed the method using influenza hemagglutinin as a model viral membrane antigen, and tested it using acetylcholine receptor (AChR) as a model membrane autoantigen. The technique involves co-culturing B cells with adherent, bioorthogonally labeled cells expressing GFP-tagged antigen, and sorting GFP-capturing, newly activated B cells. Hemagglutinin-specific B cells isolated this way from vaccinated human donors expressed elevated CD20, CD27, CD71, and CD11c, and reduced CD21, and their secreted antibodies blocked hemagglutination and neutralized viral infection. Antibodies cloned from AChR-capturing B cells derived from patients with myasthenia gravis bound specifically to the receptor on cell membrane. The approach is sensitive enough to detect antigen-specific B cells at steady state, and can be adapted for any membrane antigen.


Subject(s)
Antigens, Surface/immunology , B-Lymphocytes/immunology , Cell Separation/methods , Adult , Aged , Animals , Antigens, Surface/isolation & purification , Autoantigens/immunology , Autoantigens/isolation & purification , B-Lymphocyte Subsets/immunology , Cell Line, Tumor , Clone Cells , Epitopes, B-Lymphocyte/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Middle Aged , Myasthenia Gravis/immunology , Receptors, Cholinergic/immunology
11.
J Neuroimmunol ; 326: 19-27, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30447419

ABSTRACT

Two validated assays, a bridging ELISA and a luciferase-based bioassay, were compared for detection of anti-drug antibodies (ADA) against interferon-beta (IFN-ß) in patients with multiple sclerosis. Serum samples were tested from patients enrolled in a prospective study of 18 months. In contrast to the ELISA, when IFN-ß-specific rabbit polyclonal and human monoclonal antibodies were tested, the bioassay was the more sensitive to detect IFN-ß ADA in patients' sera. For clinical samples, selection of method of ELISA should be evaluated prior to the use of a multi-tiered approach. A titer threshold value is reported that may be used as a predictor for persistently positive neutralizing ADA.


Subject(s)
Antibodies, Neutralizing/blood , Multiple Sclerosis/blood , Neutralization Tests/methods , Biological Assay , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunologic Factors/immunology , Immunologic Factors/therapeutic use , Interferon-beta/immunology , Interferon-beta/therapeutic use , Male , Multiple Sclerosis/drug therapy
12.
J Neuropathol Exp Neurol ; 67(5): 435-48, 2008 May.
Article in English | MEDLINE | ID: mdl-18431253

ABSTRACT

Gliomas, particularly glioblastoma multiforme, perturb the blood-brain barrier and cause brain edema that contributes to morbidity and mortality. The mechanisms underlying this vasogenic edema are poorly understood. We examined the effects of cocultured primary cultured human glioblastoma cells and glioma-derived growth factors on the endothelial cell tight junction proteins claudin 1, claudin 5, occludin, and zonula occludens 1 of brain-derived microvascular endothelial cells and a human umbilical vein endothelial cell line. Cocultured glioblastoma cells and glioma-derived factors (e.g. transforming growth factor beta2) enhanced the paracellular flux of endothelial cell monolayers in conjunction with downregulation of the tight junction proteins. Neutralizing anti-transforming growth factor beta2 antibodies partially restored the barrier properties in this in vitro blood-brain barrier model. The involvement of endothelial cell-derived matrix metalloproteinases (MMPs) was demonstrated by quantitative reverse-transcriptase-polymerase chain reaction analysis and by the determination of MMP activities via zymography and fluorometry in the presence or absence of the MMP inhibitor GM6001. Occludin, claudin 1, and claudin 5 were expressed in microvascular endothelial cells in nonneoplastic brain samples but were significantly reduced in anaplastic astrocytoma and glioblastoma samples. Taken together, these in vitro and in vivo results indicate that glioma-derived factors may induce MMPs and downregulate endothelial tight junction protein and, thus, play a key role in glioma-induced impairment of the blood-brain barrier.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Endothelial Cells/metabolism , Glioblastoma/metabolism , Matrix Metalloproteinases/metabolism , Tight Junctions/metabolism , Transforming Growth Factor beta2/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/blood supply , Brain/pathology , Brain/physiopathology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/physiopathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Cells, Cultured , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Claudin-1 , Claudin-5 , Coculture Techniques , Down-Regulation/physiology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glioblastoma/pathology , Glioblastoma/physiopathology , Humans , Infant, Newborn , Membrane Proteins/metabolism , Occludin , Tight Junctions/pathology , Transforming Growth Factor beta2/pharmacology , Tumor Cells, Cultured
13.
J Neuroimmunol ; 194(1-2): 153-64, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18178259

ABSTRACT

Natalizumab, the most recently approved treatment for relapsing multiple sclerosis (MS) exerts its action through binding to alpha4 integrins. We studied longitudinally gene expression profiles in peripheral blood of MS patients, treated with natalizumab for more than 2 years. The majority of altered genes relates to immune response, signal transduction, adhesion and metabolism. Not only gene expression relevant for T lymphocytes was altered, but also genes regulating B-lymphocyte, neutrophil and erythrocyte functions. Understanding these different gene effects and their interrelationships will provide more insights into additional mechanisms of action of natalizumab and possibly allow better prediction of adverse events.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Cells/drug effects , Gene Expression Regulation/drug effects , Multiple Sclerosis/blood , Transcription, Genetic/drug effects , Adult , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Blood Cells/metabolism , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cytokines/blood , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Follow-Up Studies , Humans , Immunity/genetics , Integrin alpha4/immunology , Male , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Natalizumab , Randomized Controlled Trials as Topic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
14.
J Mol Diagn ; 9(4): 452-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17690207

ABSTRACT

We have developed a procedure for isolation of microRNA and genomic DNA in addition to total RNA from whole blood stabilized in PAXgene Blood RNA tubes. The procedure is based on automatic extraction on a BioRobot MDx and includes isolation of DNA from a fraction of the stabilized blood and recovery of small RNA species that are otherwise lost. The procedure presented here is suitable for large-scale experiments and is amenable to further automation. Procured total RNA and DNA was tested using Affymetrix Expression and single-nucleotide polymorphism GeneChips, respectively, and isolated microRNA was tested using spotted locked nucleic acid-based microarrays. We conclude that the yield and quality of total RNA, microRNA, and DNA from a single PAXgene blood RNA tube is sufficient for downstream microarray analysis.


Subject(s)
Blood Specimen Collection/methods , DNA/isolation & purification , MicroRNAs/isolation & purification , Oligonucleotide Array Sequence Analysis , Genome, Human/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Time Factors
15.
J Neuroimmunol ; 192(1-2): 198-205, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17950468

ABSTRACT

Many multiple sclerosis (MS) patients treated with interferon-beta (IFNbeta) develop anti-IFNbeta antibodies (BAbs), which can interfere with both in vitro and in vivo bioactivity of the injected cytokine. Objective of this study was to correlate these measures. Among the 256 enrolled patients, 11 (4.3%) showed a significant inhibition of the IFNbeta activity in vitro, but no measurable BAbs. As a whole, in vivo bioactivity was inhibited in 9/11 (82%) of these patients. A minority of IFNbeta treated patients have a non-antibody mediated neutralising activity, which competitively inhibits the bioactivity both in vitro and in vivo.


Subject(s)
Antibodies/therapeutic use , Interferon-beta/immunology , Interferon-beta/therapeutic use , Multiple Sclerosis/immunology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay/methods , Female , Follow-Up Studies , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression/drug effects , Humans , Immunoprecipitation , In Vitro Techniques , Male , Multiple Sclerosis/therapy , Myxovirus Resistance Proteins , Neutralization Tests , Receptors, Interferon/metabolism , Retrospective Studies
16.
J Neurol ; 254(2): 160-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17334662

ABSTRACT

OBJECTIVE: We investigated the correlation of antimyelin oligodendrocyte glycoprotein-(anti-MOG) and anti-myelin basic protein antibodies (anti-MBP) in serum of CIS patients with inflammatory signs in MRI and in CSF and, as previously suggested,the incidence of more frequent and rapid progression to clinically definite MS (CDMS). METHODS: 133CIS patients were analysed for anti-MOG and anti-MBP (Western blot). Routine CSF and cranial MRI (quantitatively and qualitatively) measures were analyzed. 55 patients had a follow-up of at least 12 months or until conversion to CDMS. RESULTS: Patients with anti-MOG and anti-MBP had an increased intrathecal IgG production and CSF white blood cell count(p = 0.048 and p = 0.036). When anti-MBP alone, or both antibodies were present the cranial MRI showed significantly more T2 lesions (p = 0.007 and p = 0.01,respectively). There was a trend for more lesion dissemination in anti-MBP positive patients (p = 0.076).Conversely, anti-MOG- and/or anti-MBP failed to predict conversion to CDMS in our follow-up group (n = 55). Only in female patients with at least one MRI lesion (n = 34) did the presence of anti-MOG correlate with more frequent (p = 0.028) and more rapid (p = 0.0209) transition to CDMS. CONCLUSIONS: Presence of anti-MOG or anti-MBP or both was not significantly associated with conversion to CDMS in our CIS cohort. However, patients with anti-MOG and anti-MBP had higher lesion load and more disseminated lesions in cranial MRI as well as higher values for CSF leucocytes and intrathecal IgG production. Our data support a correlation of anti-MOG and anti-MBP to inflammatory signs in MRI and CSF. The prognostic value of these antibodies for CDMS, however, seems to be less pronounced than previously reported.


Subject(s)
Antibodies/cerebrospinal fluid , Inflammation , Magnetic Resonance Imaging , Multiple Sclerosis , Myelin Basic Protein/immunology , Myelin-Associated Glycoprotein/immunology , Adult , Chi-Square Distribution , Female , Follow-Up Studies , Humans , Inflammation/cerebrospinal fluid , Inflammation/complications , Inflammation/pathology , Male , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Retrospective Studies , Statistics, Nonparametric
17.
BMC Biol ; 4: 15, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16749930

ABSTRACT

BACKGROUND: Pneumococcal meningitis is associated with high mortality (approximately 30%) and morbidity. Up to 50% of survivors are affected by neurological sequelae due to a wide spectrum of brain injury mainly affecting the cortex and hippocampus. Despite this significant disease burden, the genetic program that regulates the host response leading to brain damage as a consequence of bacterial meningitis is largely unknown. We used an infant rat model of pneumococcal meningitis to assess gene expression profiles in cortex and hippocampus at 22 and 44 hours after infection and in controls at 22 h after mock-infection with saline. To analyze the biological significance of the data generated by Affymetrix DNA microarrays, a bioinformatics pipeline was used combining (i) a literature-profiling algorithm to cluster genes based on the vocabulary of abstracts indexed in MEDLINE (NCBI) and (ii) the self-organizing map (SOM), a clustering technique based on covariance in gene expression kinetics. RESULTS: Among 598 genes differentially regulated (change factor > or = 1.5; p < or = 0.05), 77% were automatically assigned to one of 11 functional groups with 94% accuracy. SOM disclosed six patterns of expression kinetics. Genes associated with growth control/neuroplasticity, signal transduction, cell death/survival, cytoskeleton, and immunity were generally upregulated. In contrast, genes related to neurotransmission and lipid metabolism were transiently downregulated on the whole. The majority of the genes associated with ionic homeostasis, neurotransmission, signal transduction and lipid metabolism were differentially regulated specifically in the hippocampus. Of the cell death/survival genes found to be continuously upregulated only in hippocampus, the majority are pro-apoptotic, while those continuously upregulated only in cortex are anti-apoptotic. CONCLUSION: Temporal and spatial analysis of gene expression in experimental pneumococcal meningitis identified potential targets for therapy.


Subject(s)
Cerebral Cortex/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Meningitis, Pneumococcal/genetics , Oligonucleotide Array Sequence Analysis , Acute Disease , Algorithms , Animals , Blood-Brain Barrier , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death , Cell Survival , Cerebral Cortex/pathology , Hippocampus/pathology , Homeostasis , Meningitis, Pneumococcal/metabolism , Meningitis, Pneumococcal/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction
18.
Neurol Neuroimmunol Neuroinflamm ; 3(3): e223, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27088119

ABSTRACT

OBJECTIVES: To assess messenger RNA (mRNA) expression of POU2AF1 and Spi-B and their potential regulatory microRNAs (miRNAs) in natalizumab-treated patients with multiple sclerosis and in therapy-associated progressive multifocal leukoencephalopathy (PML). METHODS: Expression of POU2AF1/Spi-B was analyzed by using real-time reverse transcription PCR assays on isolated B/CD8(+) T lymphocytes and peripheral blood mononuclear cells (PBMCs) from cohorts of untreated and natalizumab-treated patients with and without PML. Longitudinal expression analysis was performed on CD4(+), CD8(+) T and B cells from 14 patients who interrupted natalizumab therapy for 8 weeks. The miRNA profiling was conducted in PBMCs from 5 untreated and 5 natalizumab-treated patients using low-density arrays followed by validation with single miRNAs assays in untreated and natalizumab-treated patients. RESULTS: POU2AF1 and Spi-B mRNAs were upregulated in B and CD8(+) T cells from natalizumab-treated patients, which was validated in PBMCs from different cohorts of natalizumab-treated patients with and without PML, with a noteworthy higher expression of Spi-B in patients with PML. In contrast, downregulation of POU2AF1/Spi-B expression was measured in B and CD8(+) T cells after natalizumab discontinuation. Seventeen differentially expressed miRNAs including miR-10b, a regulator of POU2AF1 mRNA, were identified in long-term natalizumab-treated patients compared with untreated ones. CONCLUSIONS: Upregulation of POU2AF1 and Spi-B, known transactivators of the JC virus, the causative agent for PML, and its association with occurrence of PML in natalizumab-treated patients, corroborates POU2AF1/Spi-B as potential biomarkers for PML risk, which merits further evaluation.

19.
PLoS One ; 11(1): e0146190, 2016.
Article in English | MEDLINE | ID: mdl-26734938

ABSTRACT

BACKGROUND: Fingolimod is a first in class oral compound approved for the treatment of relapsing-remitting multiple sclerosis (RR-MS). The aim of this study was to evaluate clinical and neuroradiological responses to fingolimod as well as the safety and tolerability in RR-MS patients in clinical practice. In addition, a panel of pro-inflammatory serum cytokines was explored as potential biomarker for treatment response. METHODS: We conducted a retrospective, non-randomized, open-label, observational study in 105 patients with RR-MS and measured cytokines in longitudinal serum samples. RESULTS: Compared to the year before fingolimod start the annualized relapse rate was reduced by 44%. Also, the percentage of patients with a worsening of the EDSS decreased. Accordingly, the fraction of patients with no evidence of disease activity (no relapse, stable EDSS, no new active lesions in MRI) increased from 11% to 38%. The efficacy and safety were comparable between highly active patients or patients with relevant comorbidities and our general patient population. CONCLUSIONS: The efficacy in reducing relapses was comparable to that observed in the phase III trials. In our cohort fingolimod was safe and efficacious irrespective of comorbidities and previous treatment.


Subject(s)
Fingolimod Hydrochloride/adverse effects , Immunosuppressive Agents/adverse effects , Multiple Sclerosis, Relapsing-Remitting/pathology , Adult , Aged , Case-Control Studies , Cytokines/blood , Dyspnea/etiology , Edema/etiology , Female , Fingolimod Hydrochloride/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Recurrence , Retrospective Studies
20.
Neurol Neuroimmunol Neuroinflamm ; 3(2): e202, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26894205

ABSTRACT

OBJECTIVE: To evaluate serum cytokine profiles for their utility to determine the heterogeneous responses to interferon (IFN)-ß treatment in patients with multiple sclerosis (MS). METHODS: Patients with relapsing-remitting MS (RRMS) or clinically isolated syndrome receiving de novo IFN-ß treatment were included in this prospective, observational study. Number of relapses and changes in disability were assessed 2 years prior to and 2 years after initiation of treatment. Sera were collected at baseline and after 3 months on therapy. Cytokine levels in sera were assessed by Luminex multiplex assays. Baseline cytokine profiles were grouped by hierarchical clustering analysis. Demographic features, changes in cytokines, and clinical outcome were then assessed in the clustered patient groups. RESULTS: A total of 157 patients were included in the study and clustered into 6 distinct subsets by baseline cytokine profiles. These subsets differed significantly in their clinical and biological response to IFN-ß therapy. Two subsets were associated with patients who responded poorly to therapy. Two other subsets, associated with a good response to therapy, showed a significant reduction in relapse rates and no worsening of disability. Each subset also had differential changes in cytokine levels after 3 months of IFN-ß treatment. CONCLUSIONS: There is heterogeneity in the immunologic pathways of the RRMS population, which correlates with IFN-ß response.

SELECTION OF CITATIONS
SEARCH DETAIL