Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Biol Chem ; 299(4): 103065, 2023 04.
Article in English | MEDLINE | ID: mdl-36841486

ABSTRACT

The peptide spanning residues 35 to 55 of the protein myelin oligodendrocyte glycoprotein (MOG) has been studied extensively in its role as a key autoantigen in the neuroinflammatory autoimmune disease multiple sclerosis. Rodents and nonhuman primate species immunized with this peptide develop a neuroinflammatory condition called experimental autoimmune encephalomyelitis, often used as a model for multiple sclerosis. Over the last decade, the role of citrullination of this antigen in the disease onset and progression has come under increased scrutiny. We recently reported on the ability of these citrullinated MOG35-55 peptides to aggregate in an amyloid-like fashion, suggesting a new potential pathogenic mechanism underlying this disease. The immunodominant region of MOG is highly conserved between species, with the only difference between the murine and human protein, a polymorphism on position 42, which is serine in mice and proline for humans. Here, we show that the biophysical and biochemical behavior we previously observed for citrullinated murine MOG35-55 is fundamentally different for human and mouse MOG35-55. The citrullinated human peptides do not show amyloid-like behavior under the conditions where the murine peptides do. Moreover, we tested the ability of these peptides to stimulate lymphocytes derived from MOG immunized marmoset monkeys. While the citrullinated murine peptides did not produce a proliferative response, one of the citrullinated human peptides did. We postulate that this unexpected difference is caused by disparate antigen processing. Taken together, our results suggest that further study on the role of citrullination in MOG-induced experimental autoimmune encephalomyelitis is necessary.


Subject(s)
Citrullination , Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Myelin-Oligodendrocyte Glycoprotein , Animals , Humans , Mice , Amyloid , Amyloidogenic Proteins , Autoantigens/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Mice, Inbred C57BL , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Myelin-Oligodendrocyte Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein/chemistry , Peptide Fragments/chemistry
3.
Clin Exp Immunol ; 183(3): 405-18, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26540618

ABSTRACT

T cells have a central pathogenic role in the aetiopathogenesis of rheumatoid arthritis (RA), and are therefore a favoured target of immunotherapy aiming at physical or functional elimination. Here we report an efficacy test of FR104, a new co-stimulation inhibitor directly targeting CD28 on T cells, in a translationally relevant model, the rhesus monkey model of collagen-induced arthritis (CIA). As a relevant comparator we used abatacept [cytotoxic T lymphocyte antigen immunoglobulin (CTLA Ig)], an antagonist of CTLA-4 binding to CD80/86 clinically approved for treatment of RA. Treatment with either compound was started at the day of CIA induction. Although FR104 previously demonstrated a higher control of T cell responses in vitro than abatacept, both compounds were equally potent in the suppression of CIA symptoms and biomarkers, such as the production of C-reactive protein (CRP) and interleukin (IL)-6 and anti-collagen type II (CII) serum antibody (IgM/IgG). However, in contrast to abatacept, FR104 showed effective suppression of CII-induced peripheral blood mononuclear cell (PBMC) proliferation. The current study demonstrates a strong potential of the new selective CD28 antagonist FR104 for treatment of RA.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , CD28 Antigens/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Abatacept/administration & dosage , Abatacept/therapeutic use , Animals , Antirheumatic Agents/isolation & purification , Antirheumatic Agents/therapeutic use , Arthritis, Experimental , Autoimmunity/drug effects , C-Reactive Protein/metabolism , CD28 Antigens/immunology , Collagen/immunology , Drug Administration Schedule , Female , Humans , Interleukin-6/blood , Lymphocyte Activation/drug effects , Macaca mulatta , Male , T-Lymphocytes/physiology , Treatment Outcome
4.
Clin Exp Immunol ; 180(1): 28-39, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25393803

ABSTRACT

Current therapies for multiple sclerosis (MS), a chronic autoimmune neuroinflammatory disease, mostly target general cell populations or immune molecules, which may lead to a compromised immune system. A more directed strategy would be to re-enforce tolerance of the autoaggressive T cells that drive tissue inflammation and injury. In this study, we have investigated whether the course of experimental autoimmune encephalomyelitis (EAE) in mice and marmosets can be altered by a potent tolerizing fusion protein. In addition, a multi-parameter immunological analysis was performed in marmosets to assess whether the treatment induces modulation of EAE-associated cellular and humoral immune reactions. The fusion protein, CTA1R9K-hMOG10-60-DD, contains a mutated cholera toxin A1 subunit (CTA1R9K), a dimer of the Ig binding D region of Staphylococcus aureus protein A (DD), and the human myelin oligodendrocyte glycoprotein (hMOG) sequence 10-60. We observed that intranasal application of CTA1R9K-hMOG10-60-DD seems to skew the immune response against myelin oligodendrocyte glycoprotein (MOG) towards a regulatory function. We show a reduced number of circulating macrophages, reduced MOG-induced expansion of mononuclear cells in peripheral blood, reduced MOG-induced production of interleukin (IL)-17A in spleen, increased MOG-induced production of IL-4 and IL-10 and an increased percentage of cells expressing programmed cell death-1 (PD-1) and CC chemokine receptor 4 (CCR4). Nevertheless, the treatment did not detectably change the EAE course and pathology. Thus, despite a detectable effect on relevant immune parameters, the fusion protein failed to influence the clinical and pathological outcome of disease. This result warrants further development and improvement of this specifically targeted tolerance inducing therapy.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunomodulation/drug effects , Multiple Sclerosis/drug therapy , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Animals , Callithrix , Cholera Toxin/genetics , Cholera Toxin/immunology , Cholera Toxin/pharmacology , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunomodulation/genetics , Mice , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/genetics , Myelin-Oligodendrocyte Glycoprotein/immunology , Organic Chemicals , Receptors, CCR4/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Staphylococcal Protein A/genetics , Staphylococcal Protein A/immunology , Staphylococcal Protein A/pharmacology
5.
J Exp Med ; 211(7): 1465-83, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24935259

ABSTRACT

Myelin oligodendrocyte glycoprotein (MOG), a constituent of central nervous system myelin, is an important autoantigen in the neuroinflammatory disease multiple sclerosis (MS). However, its function remains unknown. Here, we show that, in healthy human myelin, MOG is decorated with fucosylated N-glycans that support recognition by the C-type lectin receptor (CLR) DC-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) on microglia and DCs. The interaction of MOG with DC-SIGN in the context of simultaneous TLR4 activation resulted in enhanced IL-10 secretion and decreased T cell proliferation in a DC-SIGN-, glycosylation-, and Raf1-dependent manner. Exposure of oligodendrocytes to proinflammatory factors resulted in the down-regulation of fucosyltransferase expression, reflected by altered glycosylation at the MS lesion site. Indeed, removal of fucose on myelin reduced DC-SIGN-dependent homeostatic control, and resulted in inflammasome activation, increased T cell proliferation, and differentiation toward a Th17-prone phenotype. These data demonstrate a new role for myelin glycosylation in the control of immune homeostasis in the healthy human brain through the MOG-DC-SIGN homeostatic regulatory axis, which is comprised by inflammatory insults that affect glycosylation. This phenomenon should be considered as a basis to restore immune tolerance in MS.


Subject(s)
Brain/immunology , Cell Adhesion Molecules/immunology , Immune Tolerance/physiology , Inflammasomes/immunology , Lectins, C-Type/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Cell Surface/immunology , Th17 Cells/immunology , Animals , Brain/cytology , CHO Cells , Cell Adhesion Molecules/genetics , Cell Proliferation , Cricetinae , Cricetulus , Female , Humans , Inflammasomes/genetics , Inflammation Mediators/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Lectins, C-Type/genetics , Male , Myelin-Oligodendrocyte Glycoprotein/genetics , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/immunology , Receptors, Cell Surface/genetics , Th17 Cells/cytology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
6.
Clin Exp Immunol ; 174(1): 161-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23750720

ABSTRACT

Macaques provide important animal models in biomedical research into infectious and chronic inflammatory disease. Therefore, a proper understanding of the similarities and differences in immune function between macaques and humans is needed for adequate interpretation of the data and translation to the human situation. Dendritic cells are important as key regulators of innate and adaptive immune responses. Using a new whole blood assay we investigated functional characteristics of blood plasmacytoid dendritic cells (pDC), myeloid dendritic cells (mDC) and monocytes in rhesus macaques by studying induction of activation markers and cytokine expression upon Toll-like receptor (TLR) stimulation. In a head-to-head comparison we observed that rhesus macaque venous blood contained relatively lower numbers of pDC than human venous blood, while mDC and monocytes were present at similar percentages. In contrast to humans, pDC in rhesus macaques expressed the interleukin (IL)-12p40 subunit in response to TLR-7/8 as well as TLR-9 stimulation. Expression of IL-12p40 was confirmed by using different monoclonal antibodies and by reverse transcription-polymerase chain reaction (RT-PCR). Both in humans and rhesus macaques, TLR-4 stimulation induced IL-12p40 expression in mDC and monocytes, but not in pDC. The data show that, in contrast to humans, pDC in macaques are able to express IL-12p40, which could have consequences for evaluation of human vaccine candidates and viral infection.


Subject(s)
Dendritic Cells/immunology , Interleukin-12 Subunit p40/biosynthesis , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/blood , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/blood , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/blood , Animals , Dendritic Cells/metabolism , Humans , Interleukin-12 Subunit p40/blood , Interleukin-12 Subunit p40/genetics , Macaca mulatta
7.
Clin Exp Immunol ; 160(3): 466-78, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345979

ABSTRACT

The marked improvement of several immune-mediated inflammatory diseases during pregnancy has drawn attention to pregnancy hormones as potential therapeutics for such disorders. Low molecular weight fractions derived from the pregnancy hormone human chorionic gonadotrophin (hCG) have remarkable potent immunosuppressive effects in mouse models of diabetes and septic shock. Based on these data we have designed a set of oligopeptides related to the primary structure of hCG and tested these in models of septic shock in mice and rhesus monkeys. We demonstrate that mice exposed to lipopolysaccharide (LPS) and treated subsequently with selected tri-, tetra-, penta- and hepta-meric oligopeptides (i.e. MTR, VVC, MTRV, LQGV, AQGV, VLPALP, VLPALPQ) are protected against fatal LPS-induced septic shock. Moreover, administration of a cocktail of three selected oligopeptides (LQGV, AQGV and VLPALP) improved the pathological features markedly and nearly improved haemodynamic parameters associated with intravenous Escherichia coli-induced septic shock in rhesus monkeys. These data indicate that the designed hCG-related oligopeptides may present a potential treatment for the initial hyperdynamic phase of septic shock in humans.


Subject(s)
Chorionic Gonadotropin/pharmacology , Escherichia coli Infections/prevention & control , Escherichia coli , Oligopeptides/pharmacology , Shock, Septic/prevention & control , Amino Acid Sequence , Animals , Female , Humans , Lipopolysaccharides/toxicity , Macaca mulatta , Mice , Mice, Inbred BALB C , Pregnancy , Shock, Septic/chemically induced , Shock, Septic/microbiology
8.
Gene Ther ; 10(10): 890-901, 2003 May.
Article in English | MEDLINE | ID: mdl-12732874

ABSTRACT

Before autoimmune diseases in humans can be treated with gene therapy, the safety and efficacy of the used vectors must be tested in valid experimental models. Monkeys, such as the rhesus macaque or the common marmoset, provide such models. This publication reviews the state of the art in monkey models for rheumatoid arthritis and multiple sclerosis and the (few) gene therapy experiments that have been performed in these models.


Subject(s)
Autoimmune Diseases/therapy , Genetic Therapy/methods , Models, Animal , Primates , Animals , Arthritis/immunology , Arthritis/therapy , Autoimmune Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Vectors/administration & dosage , Haplorhini , Humans , Interleukin-4/genetics , Interleukin-4/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy
9.
J Immunol ; 167(5): 2942-9, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11509643

ABSTRACT

Inhibition of CD40-CD40 ligand interaction is a potentially effective approach for treatment of autoimmune diseases, such as multiple sclerosis. We have investigated this concept with a chimeric antagonist anti-human CD40 mAb (ch5D12) in the marmoset monkey experimental autoimmune encephalomyelitis (EAE) model. Marmosets were immunized with recombinant human myelin oligodendrocyte glycoprotein (rMOG) and treated from the day before immunization (day -1) until day 50 with either ch5D12 (5 mg/kg every 2-4 days) or placebo. On day 41 after the induction of EAE, four of four placebo-treated monkeys had developed severe clinical EAE, whereas all animals from the ch5D12-treated group were completely free of disease symptoms. High serum levels of ch5D12 associated with complete coating of CD40 on circulating B cells were found. At necropsy placebo- and ch5D12-treated animals showed similar MOG-specific lymphoproliferative responses in vitro, but ch5D12 treatment resulted in strongly reduced anti-MOG IgM Ab responses and delayed anti-MOG IgG responses. Most importantly, treatment with ch5D12 prevented intramolecular spreading of epitope recognition. Postmortem magnetic resonance imaging and immunohistologic analysis of the CNS showed a markedly reduced lesion load after ch5D12 treatment. In conclusion, the strong reduction of clinical, pathological, and radiological aspects of EAE by ch5D12 treatment in this preclinical model points to a therapeutic potential of this engineered antagonist anti-CD40 mAb for multiple sclerosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , B-Lymphocytes/immunology , CD40 Antigens/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Animals , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Callithrix , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Immunization , Lymphocyte Activation , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Recombinant Fusion Proteins/pharmacology , Time Factors
10.
J Neuroimmunol ; 119(1): 124-30, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11525809

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) induced by immunization of mice with epitopes of the proteolipid protein (PLP), a major myelin constituent, forms a useful model for the study of multiple sclerosis (MS). In addition, MS patients display PLP-specific T- and B-cell responses, suggesting that PLP reactivity is relevant to pathogenesis.Here, the generation and characterization of a panel of mouse monoclonal antibodies (Mab) against PLP139-151, the prominent encephalitogenic sequence in SJL/J mice is described. Five Mab were generated by conventional immunization of an SJL/J mouse and hybridoma generation. These Mab reacted well with the PLP139-151 peptide in ELISA and belonged to the IgG2a and IgG2b subclasses, consistent with CD4+ T helper 1-cell-supported antibody formation. The Mab also efficiently detected PLP peptide-BSA conjugates in Western blot, confirming their multi-assay applicability. The Mab were subsequently used to determine the occurrence of demyelination in brains of MS patients and marmoset monkeys with EAE. Immunohistochemistry on both paraffin and frozen sections demonstrated a homogeneous expression of PLP139-151 in normal myelin, and a complete absence in lesions containing demyelinated areas, confirming that the Mab can be used as a general myelin marker. In active demyelinating MS lesions, the Mab visualized the peptide in the cytoplasm of macrophages containing phagocytosed myelin. In conclusion, this panel of Mab against the encephalitogenic PLP139-151 epitope forms a useful tool for further study of autoantigen expression, demyelination/remyelination and the staging of lesional activity in MS patients, as well as in EAE models in distinct animal species.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Proteolipid Protein/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Peptide Fragments/metabolism , Animals , Antibodies, Monoclonal , Callithrix , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Female , Humans , Macrophages/metabolism , Mice , Mice, Inbred Strains , Reference Values
11.
Clin Exp Immunol ; 124(1): 134-41, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11359452

ABSTRACT

CIA in the rhesus monkey is an autoimmune-based polyarthritis with inflammation and erosion of synovial joints that shares various features with human rheumatoid arthritis (RA). The close phylogenetic relationship between man and rhesus monkey makes the model very suitable for preclinical safety and efficacy testing of new therapeutics with exclusive reactivity in primates. In this study we have investigated the prophylactic and therapeutic effects of a humanized monoclonal antibody (Daclizumab) against the alpha-chain of the IL-2 receptor (CD25). When Daclizumab treatment was started well after immunization but before the expected onset of CIA a significant reduction of joint-inflammation and joint-erosion was observed. A therapeutic treatment, initiated as soon as the first clinical signs of CIA were observed, proved also effective since joint-degradation was abrogated. The results of this study indicate that Daclizumab has clinical potential for the treatment of RA during periods of active inflammation and suppression of the destruction of the joint tissues.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Autoimmune Diseases/drug therapy , Immunoglobulin G/therapeutic use , Receptors, Interleukin-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Antibody Specificity , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/prevention & control , Autoantibodies/blood , Autoantibodies/immunology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , C-Reactive Protein/analysis , Collagen/immunology , Collagen/toxicity , Daclizumab , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Hydroxyproline/urine , Immunization , Macaca mulatta , Male , Receptors, Interleukin-2/immunology , Weight Loss
12.
Hum Immunol ; 62(12): 1371-81, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11756006

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease primarily affecting the central nervous system. Of the many candidate polymorphic major histocompatibility complex (MHC) and non-MHC genes contributing to disease susceptibility, including those encoding effector (cytokines and chemokines) or receptor molecules within the immune system (MHC, TCR, Ig or FcR), human leukocyte antigen (HLA) class II genes have the most significant influence. In this article we put forward the hypothesis that the influence of HLA genes on the risk to develop MS is actually the sum of multiple antigen presenting cell (APC) and T-cell interactions involving HLA class I and class II molecules. This article will also discuss that, because of the genetic and immunologic similarity to humans, autoimmune models of MS in non-human primates are the experimental models "par excellence" to test this hypothesis.


Subject(s)
Disease Models, Animal , Major Histocompatibility Complex , Multiple Sclerosis/genetics , Animals , Antigen-Presenting Cells/immunology , Autoantibodies/immunology , Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Humans , Major Histocompatibility Complex/immunology , Male , Mice , Myelin Sheath/immunology , Risk Factors , T-Lymphocytes/immunology
13.
Immunol Rev ; 183: 173-85, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11782256

ABSTRACT

The phylogenetic proximity between non-human primate species and humans is reflected by a high degree of immunological similarity. Non-human primates therefore provide important experimental models for disorders in the human population that are caused by the immune system, such as autoimmune diseases. In this paper we describe non-human primate models of multiple sclerosis, a chronic inflammatory and demyelinating disease of the human central nervous system. While reviewing data from the literature and our own research we will discuss the unique role of such models in the research of basic disease mechanisms and the development of new therapies.


Subject(s)
Callithrix , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Macaca , Multiple Sclerosis/etiology , Animals , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glycoproteins/immunology , Male , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology
14.
Clin Exp Rheumatol ; 18(5): 547-52, 2000.
Article in English | MEDLINE | ID: mdl-11072592

ABSTRACT

OBJECTIVES: To compare the activity of the CytoMegaloVirus promoter (CMV) and the Major Late promoter (MLP) in synoviocytes in vitro and in vivo. To determine the phenotype of infected cells and the induction of inflammation. To investigate the effects of the cytomegalovirus (CMV) or major late (MLP) promoter on adenovirus-mediated reporter gene transduction of synoviocytes in vitro and in vivo. METHODS: After infection with adenoviral vectors harboring CMV- and MLP-driven luciferase and lacZ genes, gene expression was examined in cultured synoviocytes and in the synovium of rhesus monkeys with collagen-induced arthritis. Immunohistochemical staining for the macrophage-marker CD68 and lacZ expression was performed. Inflammation was scored in the synovial membrane of injected and non-injected joints. RESULTS: CMV-driven reporter gene expression was found to be 6 to 10 times higher than MLP-driven gene expression in both cultured synoviocytes and monkey synovium. Both CD68 positive and CD68 negative cells were lacZ positive. Inflammation in joints injected with CMV-driven adenoviral vectors was not higher than that in MLP-driven adenoviral vectors- or non-injected joints. CONCLUSION: These experiments show that the CMV promoter induces higher gene expression in synoviocytes than the MLP promoter. Both fibroblast-like and macrophage-like synoviocytes can be infected with adenoviral vectors. No deleterious effects of the CMV-promoter driven adenoviral vectors were observed.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Promoter Regions, Genetic/physiology , Synovial Membrane/pathology , Synovial Membrane/physiopathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Arthritis/chemically induced , Arthritis/genetics , Arthritis/pathology , Cells, Cultured , Collagen , Cytomegalovirus/genetics , Female , Gene Expression , Genes, Reporter , Hyperplasia , Immunohistochemistry , Lac Operon/genetics , Macaca mulatta , Male , Reference Values
15.
J Neuroimmunol ; 110(1-2): 83-96, 2000 Oct 02.
Article in English | MEDLINE | ID: mdl-11024537

ABSTRACT

Eight rhesus monkeys with different MHC backgrounds were immunized with myelin oligodendrocyte glycoprotein (MOG). All developed severe experimental autoimmune encephalomyelitis associated with large inflammatory foci and extensive demyelination. T-cell autoreactivity to MOG was directed against three main epitopes encompassed within amino acids 4-20, 35-50 and 94-116, of which two are also immunodominant epitopes for the autoimmune T cell response to MOG in patients with MS. A strong B cell response to MOG was observed in all monkeys and major epitopes recognized were located within amino acids 4-26, 24-46 and 44-66/54-76.


Subject(s)
Central Nervous System/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Macaca mulatta , Myelin-Associated Glycoprotein/immunology , Animals , Autoantibodies/blood , Cell Lineage/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunization , Immunodominant Epitopes/immunology , Male , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myelin Proteins , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein
16.
Immunol Today ; 21(6): 290-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10825741

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) in outbred marmoset monkeys (Callithrix jacchus) is a recently developed nonhuman primate model of multiple sclerosis. Here, Bert 't Hart and colleagues compare this model to EAE in rhesus monkeys, highlighting autoimmune mechanisms in CNS inflammation and demyelination, including the role of major histocompatibility complex restriction and preclinical evaluation of innovative immunotherapies.


Subject(s)
Autoimmune Diseases/immunology , Callithrix/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Acute Disease , Animals , Antibodies, Monoclonal/therapeutic use , CD40 Antigens/immunology , Cebidae , Cercopithecidae , Chimera , Demyelinating Diseases , Desensitization, Immunologic , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Genes, MHC Class II , Histocompatibility Antigens/immunology , Immunosuppressive Agents/therapeutic use , Immunotherapy , Interferon-beta/therapeutic use , Rats , Rats, Inbred Lew , Receptors, Antigen, T-Cell/genetics , Rodentia , Rolipram/therapeutic use , Species Specificity , Twins
17.
Hum Gene Ther ; 10(7): 1139-49, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10340546

ABSTRACT

Gene transfer to synovial tissue by adenoviral vectors (Ad) was studied in vitro in cultured human synoviocytes and in vivo in seven primates with arthritis. Hyperplastic synovium was efficiently transduced with Ad.lacZ in vitro and in vivo in rhesus monkeys with collagen-induced arthritis, whereas chondrocytes were not transduced. Intraarticular injection of recombinant Ad harboring the luciferase gene showed the presence of reporter gene products only in Ad-injected joints. In addition, the feasibility of synovectomy by Ad harboring the herpes simplex virus thymidine kinase gene (tk) was studied. In vitro infection of synovium from rheumatoid arthritis patients with Ad.TK, followed by administration of ganciclovir, resulted in death of >90% of the synoviocytes. By mixing Ad.TK-infected with noninfected cells, it appeared that the presence of 10% infected synoviocytes resulted in the killing of more than 85% of the synoviocytes, demonstrating a substantial bystander effect. Intraarticular injection of Ad.TK in the knees of rhesus monkeys with arthritis, followed by treatment with ganciclovir for 14 days, resulted in increased apoptotic cell death in the synovium of Ad.TK-injected as compared with noninjected joints and ablation of the synovial lining layer. The procedure revealed no toxic side effects. These data suggest that nonsurgical synovectomy by tK gene therapy is feasible.


Subject(s)
Adenoviridae/genetics , Arthritis, Rheumatoid/therapy , Gene Transfer Techniques , Genetic Vectors , Luciferases/genetics , Synovial Membrane/metabolism , Animals , Antiviral Agents/therapeutic use , Apoptosis , Arthritis, Rheumatoid/chemically induced , Collagen/immunology , Ganciclovir/therapeutic use , Genetic Therapy , Humans , Macaca mulatta , Simplexvirus/enzymology , Synovial Membrane/cytology , Thymidine Kinase/genetics
18.
Arthritis Rheum ; 41(8): 1481-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9704648

ABSTRACT

OBJECTIVE: It has been hypothesized that asymptomatic synovitis may precede clinical manifestations of arthritis in the earliest phase of rheumatoid arthritis (RA). To obtain more insight into this disease phase, we investigated the immunohistologic features of synovial tissue (ST) from the knee joints of rhesus monkeys with induced arthritis and from RA patients with both clinically involved and clinically uninvolved knee joints. METHODS: Serial ST biopsy specimens from the knee joints of 4 rhesus monkeys that had been immunized with type II collagen and ST from 10 RA patients were investigated. Eight patients without inflammatory joint disease served as controls. RESULTS: In ST from immunized monkeys, an influx of macrophages was observed well before the occurrence of arthritis. Signs of inflammation were also demonstrated in ST from clinically uninvolved knee joints of all RA patients evaluated. The ST was characterized in particular by infiltration with macrophages and by the expression of macrophage-derived cytokines. CONCLUSION: The findings support the view that asymptomatic synovitis precedes clinically manifest arthritis in both early and established RA. This implies that the debut of RA already represents a chronic phase of the disease.


Subject(s)
Arthritis, Rheumatoid/complications , Synovitis/complications , Aged , Aged, 80 and over , Animals , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , CD3 Complex/analysis , Cytokines/metabolism , Female , Humans , Immunohistochemistry , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Macaca mulatta , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovitis/immunology , Synovitis/pathology
19.
FEBS Lett ; 409(1): 91-5, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9199510

ABSTRACT

Amino acid residues 3-15 of mycobacterial HSP60 define a dominant T-cell epitope for HLA-DR3+ve humans and Mamu-DR3+ve rhesus monkeys. Our results show that Mamu-DR3 molecules on PBMC can be efficiently loaded in vivo with the above-mentioned peptides when they are intravenously injected encapsulated in liposomes, but not in the free form. Mamu-DR3 loading is abolished by encapsulation of a nonstimulatory peptide. These results have implications for the delivery of therapeutic peptides in vivo.


Subject(s)
Histocompatibility Antigens Class II/administration & dosage , Liposomes/pharmacology , Peptides/administration & dosage , Peptides/immunology , Animals , Binding, Competitive/drug effects , Binding, Competitive/immunology , Drug Carriers , Female , Histocompatibility Antigens Class II/drug effects , Histocompatibility Antigens Class II/metabolism , Injections, Intravenous , Liposomes/administration & dosage , Lymphocyte Activation/drug effects , Macaca mulatta , Male , Peptides/metabolism , Protein Binding/drug effects , Protein Binding/immunology , T-Lymphocytes/immunology
20.
Virology ; 229(1): 175-82, 1997 Mar 03.
Article in English | MEDLINE | ID: mdl-9123859

ABSTRACT

This study aims in establishing the in vitro basis for a primate model to evaluate potential applications of H. saimiri-transformed T cells. T cell lines specific for myelin basic protein and streptolysin O were derived from rhesus monkeys and transformed to stable antigen-independent growth with strain C488 of H. saimiri. The transformed T cells from rhesus monkeys did not produce infectious virus and harbored the H. saimiri genome exclusively in an episomal form, whereas transformed T cells from the New World monkey Calltithrix jacchus released infectious virus. Transformed T cells from rhesus monkeys showed an unaltered surface expression of CD2 and CD3, of the activation markers CD25 and CD69, and of the costimulatory molecule CD80 (B7.1). Remarkably, both transformed and nontransformed T cell lines were largely double-positive for CD4 and CD8. In contrast to the parental cell lines, the transformed cells constitutively expressed major histocompatibility complex-DR antigens and were able to present antigen to each other. The transformed T cells from rhesus monkeys continued to express a functionally intact T cell receptor and responded to recognition of their antigen with enhanced proliferation and production of Th1-type cytokines. In conclusion, H. saimiri-transformed rhesus monkey T cells may open a way to primate models for adoptive immunotherapy and studies on the pathogenesis of autoaggressive T cells.


Subject(s)
Cell Transformation, Viral , Herpesvirus 2, Saimiriine/physiology , T-Lymphocytes/cytology , Animals , Antigen-Presenting Cells/immunology , CD2 Antigens/immunology , CD3 Complex/immunology , Cell Division/immunology , Cell Line , Female , Lymphocyte Activation/immunology , Macaca mulatta , Male , T-Lymphocytes/immunology , T-Lymphocytes/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...