Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
PLoS One ; 3(11): e3682, 2008.
Article in English | MEDLINE | ID: mdl-18997868

ABSTRACT

BACKGROUND: We have previously shown that vaccination with DNA encoding the encephalitogenic peptide myelin oligodendrocyte glycoprotein (MOG)(91-108) (pMOG) suppresses MOG(91-108)-induced rat Experimental Autoimmune Encephalomyelitis (EAE), a model for human Multiple Sclerosis (MS). The suppressive effect of pMOG is dependent on inclusion of CpG DNA in the plasmid backbone and is associated with early induction of Interferon (IFN)-beta. PRINCIPAL FINDINGS: In this study we examined the mechanisms underlying pMOG-induced protection. We found that in the DNA vaccinated cohort proinflammatory Interleukin (IL)-17 and IL-21 responses were dramatically reduced compared to in the control group, but that the expression of Foxp3 and Tumor Growth Factor (TGF)-beta1, which are associated with regulatory T cells, was not enhanced. Moreover, genes associated with Type I IFNs were upregulated. To delineate the role of IFN-beta in the protective mechanism we employed short interfering RNA (siRNA) to IFN-beta in the DNA vaccine. SiRNA to IFN-beta completely abrogated the protective effects of the vaccine, demonstrating that a local early elaboration of IFN-beta is important for EAE protection. IL-17 responses comparable to those in control rats developed in rats injected with the IFN-beta-silencing DNA vaccine. CONCLUSIONS: We herein demonstrate that DNA vaccination protects from proinflammatory Th17 cell responses during induction of EAE. The mechanism involves IFN-beta as IL-17 responses are rescued by silencing of IFN-beta during DNA vaccination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, DNA/immunology , Animals , Cells, Cultured , Female , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/immunology , Interferon-beta/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukins/immunology , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , RNA, Small Interfering/metabolism , Rats , Rats, Inbred Lew , Transfection , Vaccines, DNA/administration & dosage
2.
J Leukoc Biol ; 76(6): 1118-24, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15371491

ABSTRACT

Rodents typically demonstrate strain-specific susceptibilities to induced autoimmune models such as experimental arthritis and encephalomyelitis. A common feature of the local pathology of these diseases is an extensive infiltration of activated macrophages (MPhi). Different functional activation states can be induced in MPhi during innate immune activation, and it is this differential activation that might be important in susceptibility/resistance to induction or perpetuation of autoimmunity. In this study, we present an extensive, comparative analysis of the activation phenotypes of MPhi derived from autoimmune-susceptible and autoimmune-resistant rat strains to describe a cellular phenotype that defines the disease phenotype. We included investigation of receptor function, intracellular signaling pathways, cytokines, and other soluble mediators released after activation of cells using a panel of stimuli embracing many activation routes. We report that activation of MPhi from the autoimmune-susceptible strain was associated with alternative activation indicated by induction of arginase activity, a lower production of classical proinflammatory mediators, and a high production of interleukin (IL)-23, and MPhi from the autoimmune-resistant strains were associated with a higher production of proinflammatory mediators, a classical activation phenotype, and preferential induction of IL-12. These MPhi phenotypes thus reflect disparate, genetic cellular programs that define autoimmune susceptibility.


Subject(s)
Autoimmune Diseases/immunology , Genetic Predisposition to Disease/genetics , Immunity, Innate/immunology , Interleukin-12/immunology , Interleukins/immunology , Macrophages/immunology , Animals , Arginase/immunology , Autoimmune Diseases/genetics , Cell Proliferation , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Cytokines/immunology , Disease Models, Animal , Enzyme Activation/immunology , Female , Immunity, Innate/genetics , Interleukin-12/biosynthesis , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/biosynthesis , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Macrophages/metabolism , Phenotype , Rats , Receptors, Cytokine/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Species Specificity
3.
J Neuroimmunol ; 149(1-2): 66-76, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15020066

ABSTRACT

DNA vaccines encoding encephalitogenic peptides protect against subsequent development of rat experimental autoimmune encephalomyelitis (EAE) through unknown mechanisms. We investigated immune cell phenotypes at different time points after DNA vaccination with vaccine encoding myelin oligodendrocyte glycoprotein peptide 91-108 and subsequent induction of EAE. In protected rats, we observed (i) no alterations in antigen-specific Th2 or Th3 responses, (ii) reduced MHC II expression on splenocytes early after EAE induction, (iii) antigen-specific upregulation of IFNbeta upon recall stimulation and (iv) reduced IL-12Rbeta2 on lymphocytes. We suggest that the underlying mechanism of DNA vaccination is associated with immunomodulation exerted by induced IFNbeta.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Gene Expression Regulation , Interferon-beta/biosynthesis , Vaccines, DNA/administration & dosage , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Cells, Cultured , Central Nervous System/cytology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry/methods , Gene Expression Regulation/immunology , Hepatitis A Virus Cellular Receptor 2 , Humans , Interferon-beta/genetics , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lipopolysaccharides/pharmacology , Lymphocytes/immunology , Lymphocytes/metabolism , Major Histocompatibility Complex/genetics , Mice , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , NK Cell Lectin-Like Receptor Subfamily B , Peptide Fragments/immunology , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Receptors, Interleukin-12 , Receptors, Virus/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Alignment , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , Vaccination , Vaccines, DNA/immunology
4.
J Neuroimmunol ; 141(1-2): 10-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12965249

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) was induced with myelin oligodendrocyte glycoprotein (MOG(1-125)) in CD4(-/-) and CD8(-/-) DBA/1 mice. Both gene-deleted mice developed clinical signs of EAE, albeit milder than in wild-type mice, suggesting that both CD4(+) and CD8(+) cells participate in disease development. Demyelination and inflammation in the central nervous system was reduced in the absence of CD8(+) T cells. Antibody depletion of CD4(+) cells completely protected CD8(-/-) mice from MOG-induced EAE while depletion of CD8(+) cells in CD4(-/-) mice resulted in fewer EAE incidence compared to that in control antibody-treated mice. Antibody depletion of CD4(+) cells in wild-type mice protected from EAE, but not depletion of CD8(+) cells, although demyelination was reduced on removal of CD8(+) T cells. Immunization with immunodominant MOG(79-96) peptide led to EAE only in the presence of pertussis toxin (PT) in the inoculum. PT also triggered an earlier onset and more severe EAE in CD8(-/-) mice. We interpret our findings such that in an ontogenic lack of CD4(+) T cells, EAE is mediated by CD8(+) and elevated levels of alphabetaCD4(-)CD8(-) cells, and that CNS damage is partly enacted by the activity of CD8(+) T cells.


Subject(s)
CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Deletion , Animals , Autoantibodies/biosynthesis , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/pathology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/pathology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Demyelinating Diseases/prevention & control , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Incidence , Injections, Intradermal , Lymphocyte Depletion , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred DBA , Mice, Knockout , Myelin Proteins , Myelin-Associated Glycoprotein/administration & dosage , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/administration & dosage , Peptide Fragments/immunology
5.
Eur J Immunol ; 33(6): 1539-47, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12778471

ABSTRACT

To prevent an organism from developing autoimmunity the body limits the number of autoreactive cells through thymic negative selection and regulates their activity through induction of suppressor T cells. Development of antigen-specific therapies provides an interesting opportunity to imitate the body's own, often effective, method of protection. Our study demonstrates that DBA/1 mice could be protected from experimental autoimmune encephalomyelitis induced through injection of recombinant myelin oligodendrocyte glycoprotein (rMOG) when they were previously immunized intraperitoneally with rMOG adsorbed to aluminium hydroxide. This protection was associated with a decreased IFN-gamma production by rMOG-specific cells, but not a decreased proliferative response. Protection was long lasting, indicating that MOG-alum vaccination might be developed as a prophylactic therapy in multiple sclerosis.


Subject(s)
Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Myelin-Associated Glycoprotein/immunology , Vaccination , Adjuvants, Immunologic , Adsorption , Alum Compounds , Animals , Autoantigens/administration & dosage , Brain/immunology , Brain/pathology , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunoglobulin G/immunology , Immunosuppression Therapy , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred DBA , Mice, Knockout , Multiple Sclerosis , Myelin Proteins , Myelin-Associated Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein , Rats , Recombinant Proteins/immunology , Spinal Cord/immunology , Spinal Cord/pathology , Th2 Cells/immunology
6.
J Immunol ; 170(4): 1806-13, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574345

ABSTRACT

Vaccination with DNA encoding a myelin basic protein peptide suppresses Lewis rat experimental autoimmune encephalomyelitis (EAE) induced with the same peptide. Additional myelin proteins, such as myelin oligodendrocyte glycoprotein (MOG), may be important in multiple sclerosis. Here we demonstrate that DNA vaccination also suppresses MOG peptide-induced EAE. MOG(91-108) is encephalitogenic in DA rats and MHC-congenic LEW.1AV1 (RT1(av1)) and LEW.1N (RT1(n)) rats. We examined the effects of DNA vaccines encoding MOG(91-108) in tandem, with or without targeting of the hybrid gene product to IgG. In all investigated rat strains DNA vaccination suppressed clinical signs of EAE. There was no requirement for targeting the gene product to IgG, but T1-promoting CpG DNA motifs in the plasmid backbone of the construct were necessary for efficient DNA vaccination, similar to the case in DNA vaccination in myelin basic protein-induced EAE. We failed to detect any effects on ex vivo MOG-peptide-induced IFN-gamma, TNF-alpha, IL-6, IL-4, IL-10, and brain-derived neurotropic factor expression in splenocytes or CNS-derived lymphocytes. In CNS-derived lymphocytes, Fas ligand expression was down-regulated in DNA-vaccinated rats compared with controls. However, MOG-specific IgG2b responses were enhanced after DNA vaccination. The enhanced IgG2b responses together with the requirement for CpG DNA motifs in the vaccine suggest a protective mechanism involving induction of a T1-biased immune response.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunosuppressive Agents/immunology , Myelin-Associated Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Th1 Cells/immunology , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , B-Lymphocyte Subsets/immunology , Cells, Cultured , CpG Islands/immunology , DNA, Bacterial/immunology , Encephalomyelitis, Autoimmune, Experimental/mortality , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Guinea Pigs , Immunosuppressive Agents/administration & dosage , Injections, Intramuscular , Mice , Molecular Sequence Data , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/genetics , Peptide Fragments/immunology , Rats , Rats, Inbred Lew , T-Lymphocyte Subsets/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...