Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Immunol Lett ; 239: 42-59, 2021 11.
Article in English | MEDLINE | ID: mdl-34418487

ABSTRACT

The incidence of multiple sclerosis (MS) and susceptibility of animals to experimental autoimmune encephalomyelitis (EAE), the most commonly used experimental model of MS, decrease with aging. Generally, autoimmune diseases develop as the ultimate outcome of an imbalance between damaging immune responses against self and regulatory immune responses (keeping the former under control). Thus, in this review the age-related changes possibly underlying this balance were discussed. Specifically, considering the central role of T cells in MS/EAE, the impact of aging on overall functional capacity (reflecting both overall count and individual functional cell properties) of self-reactive conventional T cells (Tcons) and FoxP3+ regulatory T cells (Tregs), as the most potent immunoregulatory/suppressive cells, was analyzed, as well. The analysis encompasses three distinct compartments: thymus (the primary lymphoid organ responsible for the elimination of self-reactive T cells - negative selection and the generation of Tregs, compensating for imperfections of the negative selection), peripheral blood/lymphoid tissues ("afferent" compartment), and brain/spinal cord tissues ("target" compartment). Given that the incidence of MS and susceptibility of animals to EAE are greater in women/females than in age-matched men/males, sex as independent variable was also considered. In conclusion, with aging, sex-specific alterations in the balance of self-reactive Tcons/Tregs are likely to occur not only in the thymus/"afferent" compartment, but also in the "target" compartment, reflecting multifaceted changes in both T-cell types. Their in depth understanding is important not only for envisaging effects of aging, but also for designing interventions to slow-down aging without any adverse effect on incidence of autoimmune diseases.


Subject(s)
Aging/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Age Factors , Aged , Animals , Disease Susceptibility/immunology , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Male , Middle Aged , Multiple Sclerosis/epidemiology , Multiple Sclerosis/pathology , Prevalence , Rats , Risk Factors , Sex Factors , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/growth & development , Thymus Gland/immunology
2.
PLoS One ; 10(1): e0116566, 2015.
Article in English | MEDLINE | ID: mdl-25569428

ABSTRACT

Upregulation and/or maintenance of regulatory T cells (Tregs) during an autoimmune insult may have therapeutic efficacy in autoimmune diseases. Although several immunomodulatory drugs and molecules are available, most present significant side effects over long-term use. Cinnamon is a commonly used natural spice and flavoring material used for centuries throughout the world. Here, we have explored a novel use of cinnamon powder in protecting Tregs and treating the disease process of experimental allergic encephalomyelitis (EAE), an animal model of MS. Oral feeding of cinnamon (Cinnamonum verum) powder suppresses clinical symptoms of relapsing-remitting EAE in female PLP-TCR transgenic mice and adoptive transfer mouse model. Cinnamon also inhibited clinical symptoms of chronic EAE in male C57/BL6 mice. Dose-dependent study shows that cinnamon powder at a dose of 50 mg/kg body wt/d or higher significantly suppresses clinical symptoms of EAE in mice. Accordingly, oral administration of cinnamon also inhibited perivascular cuffing, maintained the integrity of blood-brain barrier and blood-spinal cord barrier, suppressed inflammation, normalized the expression of myelin genes, and blocked demyelination in the central nervous system of EAE mice. Interestingly, cinnamon treatment upregulated Tregs via reduction of nitric oxide production. Furthermore, we demonstrate that blocking of Tregs by neutralizing antibodies against CD25 abrogates cinnamon-mediated protection of EAE. Taken together, our results suggest that oral administration of cinnamon powder may be beneficial in MS patients and that no other existing anti-MS therapies could be so economical and trouble-free as this approach.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Plant Extracts/therapeutic use , Administration, Oral , Animals , Blood-Brain Barrier/metabolism , Cinnamomum zeylanicum/chemistry , Cinnamomum zeylanicum/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Incidence , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/immunology , Myelin Proteolipid Protein/genetics , Myelin Sheath/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Receptors, Antigen, T-Cell/genetics , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
3.
Brain Behav Immun ; 33: 33-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23643646

ABSTRACT

Epidemiological studies have associated infection during pregnancy with increased risk of neurodevelopmental disorders in children, which is modeled in rodents by stimulating the immune system of pregnant dams with microorganisms or their mimics, such as poly(I:C) or LPS. In two prenatal mouse models, we show that in utero exposure of the fetus to cytokines/inflammatory mediators elicited by maternal immune stimulation with poly(I:C) yields offspring that exhibit a proinflammatory phenotype due to alterations in developmental programming of their immune system. Changes in the innate and adaptive immune elements of these pro-inflammatory offspring result in more robust responses following exposure to immune stimuli than those observed in control offspring from PBS-injected pregnant dams. In the first model, offspring from poly(I:C)-injected immunologically naïve dams showed heightened cellular and cytokine responses 4 h after injection of zymosan, a TLR2 agonist. In the second model, using dams with immunological memory, poly(I:C) injection during pregnancy produced offspring that showed preferential differentiation toward Th17 cell development, earlier onset of clinical symptoms of EAE, and more severe neurological deficits following immunization with MOG35-55. Such "fetal programming" in offspring from poly(I:C)-injected dams not only persists into neonatal and adult life, but also can have profound consequences on health and disease.


Subject(s)
Immunomodulation/immunology , Immunophenotyping , Mothers , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Illness Behavior/physiology , Injections, Intraperitoneal , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Poly I-C/administration & dosage , Poly I-C/adverse effects , Pregnancy , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology , Zymosan/administration & dosage
4.
Immunol Cell Biol ; 87(7): 534-45, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19488061

ABSTRACT

The administration of Th2 cytokines or immune deviation to a Th2 phenotypic response has been shown to protect against the autoimmune pathology of experimental autoimmune encephalomyelitis (EAE). To better understand the function of Th2 cytokines in the induction stage of EAE in the absence of an overt Th2 response, we immunized IL-4 receptor alpha-deficient (IL-4Ralpha(-/-)) mice, which are unable to respond to either IL-4 or IL-13. Contrary to expectations, mice lacking IL-4Ralpha had a lower incidence of EAE and a delayed onset compared to WT BALB/c mice; however, this delay did not correlate to an alteration in the Th1/Th17 cytokine balance. Instead, IL-4Ralpha-responsive macrophages were essential promoters of disease as macrophage-specific IL-4Ralpha-deficient (LysM(cre)IL-4Ralpha(-/lox)) mice were protected from EAE. The protection afforded by IL-4Ralpha-deficiency was not due to IL-10-, IFN-gamma-, NO- or IDO-mediated suppression of T-cell responses but was dependent upon the presence of regulatory T cells (Tregs). This investigation highlights the importance of macrophages and Tregs in regulating central nervous system inflammation and demonstrates that macrophages activated in the absence of Th2 cytokines can promote disease suppression by Tregs.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Macrophages/physiology , Macrophages/transplantation , Receptors, Cell Surface/genetics , T-Lymphocytes, Regulatory/physiology , Adoptive Transfer/methods , Age of Onset , Animals , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Incidence , Lymph Nodes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenotype , Receptors, Cell Surface/metabolism , Signal Transduction/genetics , Spleen/metabolism
5.
J Immunol ; 182(9): 5268-75, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380773

ABSTRACT

Experimental autoimmune encephalomyelitis is induced in B10.PL (H-2(u)) mice by immunization with the immunodominant N-terminal epitope of myelin basic protein, Ac1-9. In the present study, we show that the site of immunization impacts disease incidence and severity. This effect is more marked in female mice than in males. Although immunization in the flanks is effective in eliciting disease, delivery of Ag in the footpad and tailbase results in poor induction. Analyses of the immune responses in female mice following different immunization regimens indicates that resistance to disease is accompanied by higher levels of IFN-gamma and CD11b(+)Gr-1(int) myeloid cells. Such myeloid cells are known to have a suppressive function, and consistent with this knowledge, blockade of IFN-gamma results in increased disease activity and decreased levels of splenic CD11b(+)Gr-1(int) cells. Conversely, injection of adjuvants (CFA or Pam(3)CSK(4)) in the footpad decreases experimental autoimmune encephalomyelitis incidence and severity. Our study indicates that the site of immunization can impact the magnitude of the ensuing inflammatory response, and that at a certain threshold a protective, regulatory circuit can be elicited.


Subject(s)
Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunity, Innate , Inflammation Mediators/physiology , Interferon-gamma/physiology , Myelin Basic Protein/immunology , Peptide Fragments/immunology , Vaccination/methods , Animals , Autoantigens/administration & dosage , CD11b Antigen/biosynthesis , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Incidence , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/administration & dosage , Myeloid Cells/immunology , Myeloid Cells/metabolism , Peptide Fragments/administration & dosage , Receptors, Chemokine/biosynthesis , Severity of Illness Index
6.
J Immunol ; 180(4): 2679-85, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250480

ABSTRACT

Females tend to have stronger Th1-mediated immune responses and are more prone to develop autoimmune diseases, including multiple sclerosis. Macrophages are major effector cells capable of mediating or modulating immune responses in experimental autoimmune encephalomyelitis (EAE). IL-13 and estrogen have opposing roles on macrophages (the former enhancing and the latter inhibiting) in terms of MHC class II (MHC II) up-regulation and, thus, these factors might influence susceptibility to EAE differently in females vs males. In accordance with this hypothesis, females lacking IL-13 displayed lower incidence and milder EAE disease severity than males after immunization with myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide/CFA/pertussis toxin. Female IL-13 knockout (KO) mice with EAE consistently had reduced infiltration of CD11b(+) macrophages in the CNS along with significantly reduced expression of MHC II on these cells. Impaired MHC II expression was further corroborated upon LPS stimulation of female but not male bone marrow-derived CD11b(+) macrophages from IL-13KO mice, with restored expression after IL-13 pretreatment of female but not male macrophages. APCs from IL-13KO females induced less proliferation by MOG-35-55-reactive T cells, and splenocytes from MOG peptide-immunized females had lower expression of IL-12, IFN-gamma, MIP-2, and IFN-gamma-inducible protein 10 than males. In contrast, these splenocytes had higher expression of anti-inflammatory factors, IL-10, TGF-beta1, and FoxP3, a cytokine pattern typical of regulatory type II monocytes. These data suggest that the difference in EAE susceptibility in females is strongly influenced by gender-specific proinflammatory effects of IL-13, mediated in part through up-regulation of Th1-inducing cytokines and MHC II on CD11b(+) macrophages.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Genetic Predisposition to Disease , Interleukin-13/physiology , Sex Characteristics , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Female , Incidence , Interleukin-13/deficiency , Interleukin-13/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
J Immunol ; 174(2): 918-24, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15634914

ABSTRACT

To investigate effects of a 16.8-Mb region on rat chromosome 4q42-43 on encephalomyelitis, we performed a high-resolution mapping using a 10th generation advanced intercross line between the susceptible DA strain and the MHC identical but resistant PVG.1AV1 strain. Clinical signs of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE) developed in 29% of 772 F(10) rats. Three regions controlling disease, Eae20, Eae21, and Eae22, were mapped using 15 microsatellite markers spanning 16.8 Mb. Eae20 was a major genetic determinant within the region whereas Eae21 modified disease severity. Eae22 was identified as an epistatic region because it only displayed an effect together with Piebald Virol Glaxo (PVG) alleles on Eae20. Disease down-regulation by PVG alleles in the telomeric part of Eae20 was also demonstrated in DA rats made congenic for a approximately 1.44-Mb chromosomal region from PVG. As the region containing Eae20-Eae22 also regulates arthritis, together with the fact that the syntenic mouse 6F(2)-F(3) region regulates experimental lupus and diabetes, and the syntenic human 12p13.31-13.2 region regulates multiple sclerosis and rheumatoid arthritis, the present data point to genes that control several inflammatory diseases. The pairscan analyses of interaction, which here identified Eae22, are novel in the encephalomyelitis field and of importance in the design of further studies of this region in other diseases and species. The limited number of genes identified in Eae20, Eae21, and Eae22 enables focused examination of their relevance in mechanistic animal studies and screening of their association to human diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Epistasis, Genetic , Physical Chromosome Mapping/methods , Quantitative Trait Loci/immunology , Animals , Crosses, Genetic , Down-Regulation/genetics , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Female , Genetic Linkage/immunology , Genetic Predisposition to Disease , Genotype , Incidence , Male , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Phenotype , Rats , Rats, Inbred Strains , Severity of Illness Index
8.
J Neuroimmunol ; 150(1-2): 59-69, 2004 May.
Article in English | MEDLINE | ID: mdl-15081249

ABSTRACT

Multiple sclerosis (MS) is more prevalent in women than men. We evaluated seven different mouse strains commonly used in the study of autoimmune diseases, for sex differences in the disease course of experimental autoimmune encephalomyelitis (EAE). Greater severity of EAE was observed in the female SJL immunized with two different peptides of myelin proteolipid protein (PLP) and myelin oligodendrocyte glycoprotein (MOG) as well as in the female ASW relative to males. Female NZW mice showed a greater incidence of EAE than males. However, male B10.PL and PL/J mice showed more severe disease than females. No sex differences were noted in the C57BL/6 or NOD strains.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Sex Characteristics , Amino Acid Sequence , Animals , Castration , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Incidence , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred NZB , Mice, Inbred Strains , Molecular Sequence Data , Ovariectomy , Severity of Illness Index , Species Specificity
9.
J Immunol ; 170(9): 4776-84, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12707359

ABSTRACT

Since the basic mechanisms behind the beneficial effects of IFN-beta in multiple sclerosis (MS) patients are still obscure, here we have investigated the effects of IFN-beta gene disruption on the commonly used animal model for MS, experimental autoimmune encephalomyelitis (EAE). We show that IFN-beta knockout (KO) mice are more susceptible to EAE than their wild-type (wt) littermates; they develop more severe and chronic neurological symptoms with more extensive CNS inflammation and demyelination. However, there was no discrepancy observed between wt and KO mice regarding the capacity of T cells to proliferate or produce IFN-gamma in response to recall Ag. Consequently, we addressed the effect of IFN-beta on encephalitogenic T cell development and the disease initiation phase by passive transfer of autoreactive T cells from KO or wt littermates to both groups of mice. Interestingly, IFN-beta KO mice acquired a higher incidence and augmented EAE regardless of the source of T cells. This shows that the anti-inflammatory effect of endogenous IFN-beta is predominantly exerted on the effector phase of the disease. Histopathological investigations of CNS in the effector phase revealed an extensive microglia activation and TNF-alpha production in IFN-beta KO mice; this was virtually absent in wt littermates. This coincided with an increase in effector functions of T cells in IFN-beta KO mice, as measured by IFN-gamma and IL-4 production. We suggest that lack of endogenous IFN-beta in CNS leads to augmented microglia activation, resulting in a sustained inflammation, cytokine production, and tissue damage with consequent chronic neurological deficits.


Subject(s)
Adjuvants, Immunologic/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Deletion , Interferon-beta/deficiency , Interferon-beta/genetics , Adoptive Transfer , Animals , Autoantibodies/biosynthesis , Autoantigens/immunology , Cells, Cultured , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Genetic Predisposition to Disease , Immunophenotyping , Incidence , Inflammation/genetics , Inflammation/immunology , Interferon-beta/biosynthesis , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/immunology , Myelin Sheath/pathology , Peptide Fragments/immunology , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
10.
J Immunol ; 170(4): 1630-4, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574324

ABSTRACT

Mast cell-deficient mice (W/W(v)) exhibit significantly reduced severity of experimental allergic encephalomyelitis (EAE), a murine model of multiple sclerosis. In this study, the contribution of FcR-mediated mast cell activation to disease was examined. W/W(v) mice were reconstituted i.v. with bone marrow-derived mast cells (BMMCs) from wild-type mice or those lacking functional FcRs. Eight weeks later, EAE was induced by immunization with the myelin oligodendrocyte glycoprotein 35-55 peptide. Disease scores were analyzed in reconstituted mice and compared with age-matched W/W(v) mice and wild-type littermates. Mice reconstituted with FcRgamma(-/-) BMMCs or FcgammaRIII(-/-) BMMCs exhibited less severe clinical symptoms similar to W/W(v) controls, while reconstitution with FcRIIB(-/-) BMMCs resulted in disease significantly more severe than wild-type controls. Notably, mice reconstituted with FcgammaRIII(-/-) BMMC exhibit a relapsing-remitting course of disease. These data demonstrate that both activating and inhibitory FcRs expressed on mast cells influence the course of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Mast Cells/immunology , Mast Cells/metabolism , Receptors, Fc/physiology , Amino Acid Sequence , Animals , Autoantibodies/biosynthesis , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes/immunology , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Incidence , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Receptors, Fc/biosynthesis , Receptors, Fc/deficiency , Receptors, Fc/genetics , Severity of Illness Index
11.
J Immunol ; 170(2): 1019-26, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12517969

ABSTRACT

The B10.RIII mouse strain (H-2(r)) develops chronic experimental autoimmune encephalomyelitis (EAE) upon immunization with the myelin basic protein 89-101 peptide. EAE was induced and studied in a backcross between B10.RIII and the EAE-resistant RIIIS/J strain (H-2(r)), and a complete genome scan with microsatellite markers was performed. Five loci were significantly linked to different traits and clinical subtypes of EAE on chromosomes 1, 5, 11, 15, and 16, three of the loci having sex specificity. The quantitative trait locus on chromosome 15 partly overlapped with the Eae2 locus, previously identified in crosses between the B10.RIII and RIIIS/J mouse strains. The loci on chromosomes 11 and 16 overlapped with Eae loci identified in other mouse crosses. By analyzing the backcross animals for lymphocyte phenotypes, the proportion of B and T cells in addition to the levels of CD4(+)CD8(-) and CD4(-)CD8(+) T cells and the CD4(+)/CD8(+) ratio in spleen were linked to different loci on chromosomes 1, 2, 3, 5, 6, 11, and 15. On chromosome 16, we found significant linkage to spleen cell proliferation. Several linkages overlapped with the quantitative trait loci for disease phenotypes. The identification of subphenotypes that are linked to the same loci as disease traits could be most useful in the search for candidate genes and biological pathways involved in the pathological process.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphocyte Subsets/immunology , Quantitative Trait Loci/immunology , Animals , Crosses, Genetic , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Genetic Linkage/immunology , Genotype , Immunophenotyping , Incidence , Lymphocyte Count , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains
13.
J Immunol ; 165(10): 5932-7, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11067955

ABSTRACT

Major concern has emerged about the possible long term adverse effects of glucocorticoid treatment, which is frequently used for the prevention of chronic lung disease in preterm infants. Here we show that neonatal glucocorticoid treatment of rats increases the severity (p< or = 0.01) and incidence (p< or =0.01) of the inflammatory autoimmune disease experimental autoimmune encephalomyelitis in adult life. In search of possible mechanisms responsible for the increased susceptibility to experimental autoimmune encephalomyelitis, we investigated the reactivity of the hypothalamo-pituitary-adrenal axis and of immune cells in adult rats after neonatal glucocorticoid treatment. We observed that neonatal glucocorticoid treatment reduces the corticosterone response after an LPS challenge in adult rats (p< or =0.001). Interestingly, LPS-stimulated macrophages of glucocorticoid-treated rats produce less TNF-alpha and IL-1beta in adult life than control rats (p<0.05). In addition, splenocytes obtained from adult rats express increased mRNA levels of the proinflammatory cytokines IFN-gamma (p<0.01) and TNF-beta (p<0.05) after neonatal glucocorticoid treatment. Apparently, neonatal glucocorticoid treatment has permanent programming effects on endocrine as well as immune functioning in adult life. In view of the frequent clinical application of glucocorticoids to preterm infants, our data demonstrate that neonatal glucocorticoid treatment may be a risk factor for the development of (auto)immune disease in man.


Subject(s)
Aging/immunology , Animals, Newborn/immunology , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Aging/blood , Animals , Animals, Newborn/growth & development , Body Weight/drug effects , Cells, Cultured , Corticosterone/antagonists & inhibitors , Corticosterone/blood , Cytokines/biosynthesis , Cytokines/genetics , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Incidence , Injections, Intraperitoneal , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Activation/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Severity of Illness Index , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Neurol Sci ; 171(1): 60-4, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10567051

ABSTRACT

Blockade of the CD40 ligand (CD40L)-CD40 interaction suppresses experimental autoimmune encephalomyelitis (EAE). Since this interaction induces IL-12, an essential cytokine for EAE induction, we hypothesized that CD40L blockade may suppress EAE through IL-12 inhibition. Here we show that exogenous IL-12 abolishes the ability of anti-CD40L monoclonal antibodies to prevent EAE. Anti-IL-12 antibodies prevent this reversal and protect from EAE. These results show that IL-12 is sufficient to overcome CD40L blockade and suggest that, of the multiple consequences of the CD40L-CD40 interaction, IL-12 induction is an essential one for induction of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunosuppression Therapy , Interleukin-12/immunology , Interleukin-12/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Recombinant Proteins/adverse effects , Animals , Antibodies, Monoclonal/pharmacology , CD40 Antigens/metabolism , CD40 Ligand , Crosses, Genetic , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Guinea Pigs , Immunity, Cellular , Incidence , Interleukin-12/administration & dosage , Ligands , Membrane Glycoproteins/immunology , Mice , Mice, Inbred Strains , Recombinant Proteins/administration & dosage , Spinal Cord/immunology
15.
J Immunol ; 161(4): 1860-7, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9712054

ABSTRACT

Experimental allergic encephalomyelitis (EAE), the principal animal model of multiple sclerosis, is a genetically determined phenotype. In this study, analyses of the cumulative disease frequencies in parental, F1 hybrid, and F2 mice, derived from the EAE-susceptible SJL/J strain and the EAE-resistant B10.S/DvTe strain, confirmed that susceptibility to EAE is not inherited as a simple Mendelian trait. Whole genome scanning, using 150 informative microsatellite markers and a panel of 291 affected and 390 unaffected F2 progeny, revealed significant linkage of EAE susceptibility to marker loci on chromosomes 7 (eae4) and 17, distal to H2 (eae5). Quantitative trait loci for EAE severity, duration, and onset were identified on chromosomes 11 (eae6, and eae7), 2 (eae8), 9 (eae9), and 3 (eae10). While each locus reported in this study is important in susceptibility or disease course, interactions between marker loci were not statistically significant in models of genetic control. One locus, eae7, colocalizes to the same region of chromosome II as Orch3 and Idd4, susceptibility loci in autoimmune orchitis and insulin-dependent diabetes mellitus, respectively. Importantly, eae5 and eae7 are syntenic with human chromosomes 6p21 and 17q22, respectively, two regions of potential significance recently identified in human multiple sclerosis genome scans.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Quantitative Trait, Heritable , Animals , Chromosome Mapping , Crosses, Genetic , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Female , Genetic Markers/immunology , Humans , Incidence , Linear Models , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology
17.
J Neuroimmunol ; 42(1): 23-32, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8093702

ABSTRACT

SJL/J mice are highly susceptible to actively induced experimental allergic encephalomyelitis (EAE), whereas B10.S mice are resistant. However, both strains share the H-2s haplotype. We have previously shown that the relative susceptibility of SJL/J and B10.S mice to acute EAE correlates, respectively, with high and low responsiveness to myelin basic protein (MBP), as determined by cloning and limiting dilution analysis of in vitro T cell proliferation. Here, we have investigated the ability of SJL/J and B10.S mice to generate EAE-effector T cells in vivo. We have developed a new mouse strain, B10.S Thy 1.1, that differs at the Thy 1 locus from SJL/J and B10.S mice (both Thy 1.2) but has the same MHC and resistance pattern to EAE as do B10.S mice. Using radiation bone marrow chimeras formed between SJL/J and B10.S Thy 1.1 mice, we have shown that a population of radiosensitive prethymic cells in SJL/J bone marrow has an intrinsic potential to generate EAE-effector T cells, whereas that in B10.S Thy 1.1 bone marrow does not. This lack of detectable EAE effector cells in B10.S Thy 1.1 mice does not appear to be due to the generation of suppressor T cells or to a defect in antigen-presenting cells. Moreover, the potential of SJL/J bone marrow to generate EAE-effector T cells is not inhibited by the concomitant presence of B10.S Thy 1.1 bone marrow cells, thymocytes or dendritic cells in mixed chimeras. Hence, the relative susceptibility of SJL/J and B10.S mice to EAE appears to be directly related to the respective responder status of their T cells to MBP, as evidenced by their ability (or inability) to generate EAE-effector T cells. This high and low responder status appears in turn to be linked to non-MHC background genes, although this has not been established formally.


Subject(s)
Bone Marrow Cells , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/cytology , Animals , Antigens, Surface/pharmacology , Bone Marrow/radiation effects , Brain/pathology , Cell Division , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Female , Immunity, Innate , Incidence , Male , Membrane Glycoproteins/pharmacology , Mice , Radiation Chimera/immunology , Spinal Cord/pathology , Thy-1 Antigens
18.
Isr J Med Sci ; 27(7): 365-8, 1991 Jul.
Article in English | MEDLINE | ID: mdl-2071371

ABSTRACT

Glucocorticoids are frequently employed as immunosuppressive agents. Following clinical reports of occurrence and exacerbation of neurological immune-mediated conditions in patients receiving steroids, we studied the course of experimental allergic encephalomyelitis (EAE) in rats receiving methylprednisolone prior to and during disease induction and after the appearance of clinical signs. High methylprednisolone dose given prior to EAE induction significantly increased disease duration and the number of days at maximal disability. When the drug was given prior to and during the induction phase, both moderate and high doses exacerbated the EAE course. In contrast, treatment with methylprednisolone after the onset of clinical disease had a markedly beneficial effect. Thus, time of initiation, dosage and duration of treatment may differentially affect the course of EAE. Moreover, the natural course of a neurological immune-mediated condition may be worsened when it is triggered while the patient is under steroid treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Methylprednisolone/adverse effects , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Incidence , Methylprednisolone/administration & dosage , Rats , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...