Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cells ; 8(8)2019 07 29.
Article in English | MEDLINE | ID: mdl-31362466

ABSTRACT

NK cells (natural killer cells) being a part of the innate immune system have been shown to be involved in immunoregulation of autoimmune diseases. Previously we have shown that HINT1/Hsp70 treatment induced regulatory NK cells ameliorating experimental autoimmune encephalomyelitis (EAE) course and CD4+ T cells proliferation. NK cells were isolated from mice treated with HINT1/Hsp70 and co-cultured with proteolipid protein (PLP)-stimulated CD4+ T cells isolated from EAE mice. Cell proliferation was assessed by thymidine uptake, cytotoxicity by lactate dehydrogenase (LDH) release assay and fluorescence activated cell sorting (FACS) analysis, protein expression by Western blot, mRNA by quantitative RT-PCR. Gene related to anergy in lymphocytes (GRAIL) expression was downregulated by specific siRNA and GRAIL overexpression was induced by pcDNA-GRAIL transfection. HINT1/Hsp70 pretreatment of EAE SJL/J mice ameliorated EAE course, suppressed PLP-induced T cell proliferation by enhancing T cell expression of GRAIL as GRAIL downregulation restored T cell proliferation. HINT1/Hsp70 treatment induced immunoregulatory NK cells which inhibited PLP-stimulated T cell proliferation not depending on T cell necrosis and apoptosis. This immunoregulatory NK cell function depended on NK cell expression of GRAIL as GRAIL downregulation diminished inhibition of NK cell suppression of T cell proliferation. Similarly GRAIL overexpression in NK cells induced their regulatory function. HINT1/Hsp70 treatment generated regulatory NK cells characterized by expression of GRAIL.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , HSP70 Heat-Shock Proteins/administration & dosage , Killer Cells, Natural/cytology , Nerve Tissue Proteins/administration & dosage , Ubiquitin-Protein Ligases/genetics , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Clonal Anergy , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/etiology , Female , HSP70 Heat-Shock Proteins/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Mice , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/metabolism , Nerve Tissue Proteins/pharmacology , Proteolipids/pharmacology , Ubiquitin-Protein Ligases/metabolism
2.
Bioorg Med Chem ; 26(9): 2221-2228, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29681483

ABSTRACT

We report the novel synthesis of cyclic PLP139-151 (cPLP) and its application in SJL/J mice to study its encephalitogenic effects. Our results indicate that the cPLP analog is minimally encephalitogenic when administered to induce experimental autoimmune encephalomyelitis (low disease burden, minimal inflammatory, demyelinating and axonopathic pathology compared to its linear counterpart). Proliferation assays confirmed the low stimulatory potential of the cPLP compared to linPLP (2.5-fold lower proliferation) as well as inducing lower antibody responses. Molecular modeling showed a completely different TCR recognition profile of cPLP in regard to linPLP, where H147 replaces W144 and F151-K150 replace H147 as TCR contacts, which may explain the difference on each peptide's response.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Peptides, Cyclic/immunology , Amino Acid Sequence , Animals , Cyclization , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Epitopes/adverse effects , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Female , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Mice, Inbred Strains , Molecular Docking Simulation , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/chemistry , Myelin Proteolipid Protein/metabolism , Peptide Fragments/adverse effects , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptides, Cyclic/adverse effects , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Protein Binding , Protein Conformation , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Spinal Cord/pathology , T-Lymphocytes/metabolism
3.
J Neuroimmunol ; 313: 77-81, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29153612

ABSTRACT

In patients with multiple sclerosis, the selective serotonin reuptake inhibitor, fluoxetine, resulted in less acute disease activity. We tested the immune modulating effects of fluoxetine in a mouse model of multiple sclerosis, i.e. experimental autoimmune encephalomyelitis (EAE). We show that fluoxetine delayed the onset of disease and reduced clinical paralysis in mice with established disease. Fluoxetine had abrogating effects on proliferation of immune cells and inflammatory cytokine production by both antigen-presenting cells and T cells. Specifically, in CD4 T cells, fluoxetine increased Fas-induced apoptosis. We conclude that fluoxetine possesses immune-modulating effects resulting in the amelioration of symptoms in EAE.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Fluoxetine/therapeutic use , Animals , Apoptosis , CD11b Antigen/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Freund's Adjuvant/adverse effects , Freund's Adjuvant/immunology , Macrophages/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Transgenic , Myelin Basic Protein/genetics , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/immunology , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , fas Receptor/metabolism
4.
Autoimmunity ; 48(4): 208-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25427822

ABSTRACT

The regulation of cytokine expression by immune deviation from a pro-inflammatory to anti-inflammatory or "regulatory" milieu is crucial to the prevention of permanent central nervous system (CNS) damage in neuroinflammation. Earlier studies in the murine experimental autoimmune encephalomyelitis (EAE) model pointed to an anti-inflammatory role for the Th2 cytokine, IL-4, which was not confirmed in IL-4Rα-deficient mice (IL-4Rα(-/-)). To examine the pathological consequences of loss of responsiveness to Th2 cytokines, we compared lesion evolution in IL-4Rα(-/-) and wild type (WT) BALB/c mice immunized with PLP180-199 and investigated how altering the magnitude of the antigen-specific autoimmune response in this model affected the pathology. We found that while changing the magnitude of the peripheral antigen-specific response differentially affected the incidence of clinical disease in WT BALB/c relative to IL-4Rα(-/-) mice, the differences in incidence did not correlate to differences in pro-inflammatory cytokine production. Additionally, although only approximately 75% of WT mice developed clinical disease, lesions were observed in 100% of the mice, principally in the cerebellum, mid-brain and cerebral hemispheres, and lesion load increased with increasing pro-inflammatory cytokine production. Despite being resistant to disease induction with increasing pro-inflammatory cytokine production, lesion incidence in IL-4Rα-deficient animals was equal to their WT counterparts. However, lesion severity in IL-4Rα-deficient animals was preferentially reduced in the mid-brain and cerebral hemispheres. From these studies, we conclude that signaling through IL-4Rα has little effect on regulating the peripheral pro-inflammatory cytokine profile in this EAE variant but has distinct effects on the determination of lesion topography.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Deletion , Genetic Predisposition to Disease , Receptors, Cell Surface/genetics , Animals , Antigens/immunology , Brain/immunology , Brain/metabolism , Brain/pathology , Cytokines/biosynthesis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutation , Myelin Proteolipid Protein/adverse effects , Optic Nerve/immunology , Optic Nerve/metabolism , Optic Nerve/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
5.
J Autoimmun ; 38(4): 344-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22459490

ABSTRACT

Previously we showed that transgenic mice expressing human HLA-DR3 gene are susceptible to PLP(91-110) induced experimental autoimmune encephalomyelitis (EAE) and can serve as an animal model of multiple sclerosis (MS). HLA-DR3 mice with EAE showed increased number of CD8 T cells indicating their important role in disease pathogenesis. The role of CD8 T cells in MS, an inflammatory demyelinating disease of CNS, has been enigmatic as it has been assigned both regulatory and pathogenic roles. Therefore, to evaluate the role of CD8 T cells, we generated CD8 deficient HLA-DR3 transgenic mice (DR3.CD8(-/-)). Immunization with PLP(91-110) led to more severe EAE in DR3.CD8(-/-) mice compared to HLA-DR3 mice indicating a regulatory role for CD8 T cells. Interestingly, DR3.CD8(-/-) mice with EAE showed decreased CNS pathology compared to DR3 mice thus suggesting a pathogenic role for CD8 T cells. We show that these two subsets of CD8 T cells can be differentiated based on the surface expression of CD122 (IL-2 Rß chain). CD8 T cells expressing CD122 (CD8+CD122+) play a regulatory role while CD8+CD122- T cells act as a pathogenic subset. CD122 expressing CD8 T cells are the regulatory subset of CD8 T cells and regulate the encephalitogenic CD4 T cells through direct modulation of antigen presenting cells and/or through the release of immunoregulatory cytokines such as IL-10, IFNγ and TGFß. We also showed that adoptive transfer of CD8CD122- T cells caused increased spinal cord demyelination indicating that these are pathogenic subset of CD8 T cells. Our study suggests that CD8+ T cells play both regulatory as well as pathogenic role in disease pathogenesis of EAE. A better understanding of these subsets could aid in designing novel therapy for MS patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , HLA-DR3 Antigen/genetics , Myelin Proteolipid Protein/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Central Nervous System/immunology , Central Nervous System/pathology , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , HLA-DR3 Antigen/immunology , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/chemistry , Neuroglia/immunology
6.
PLoS One ; 6(10): e25721, 2011.
Article in English | MEDLINE | ID: mdl-21998684

ABSTRACT

We report here the design, development and performance of a novel formulation of liposome- encapsulated glucocorticoids (GCs). A highly efficient (>90%) and stable GC encapsulation was obtained based on a transmembrane calcium acetate gradient driving the active accumulation of an amphipathic weak acid GC pro-drug into the intraliposome aqueous compartment, where it forms a GC-calcium precipitate. We demonstrate fabrication principles that derive from the physicochemical properties of the GC and the liposomal lipids, which play a crucial role in GC release rate and kinetics. These principles allow fabrication of formulations that exhibit either a fast, second-order (t(1/2) ~1 h), or a slow, zero-order release rate (t(1/2) ~ 50 h) kinetics. A high therapeutic efficacy was found in murine models of experimental autoimmune encephalomyelitis (EAE) and hematological malignancies.


Subject(s)
Chemistry, Pharmaceutical/methods , Glucocorticoids/administration & dosage , Glucocorticoids/chemistry , Nanostructures/chemistry , Animals , Capsules , Chemical Phenomena , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions , Leukemia/drug therapy , Liposomes , Lymphoma, T-Cell/drug therapy , Methylprednisolone Hemisuccinate/administration & dosage , Methylprednisolone Hemisuccinate/chemistry , Methylprednisolone Hemisuccinate/pharmacokinetics , Methylprednisolone Hemisuccinate/therapeutic use , Mice , Multiple Sclerosis/drug therapy , Myelin Proteolipid Protein/adverse effects , Solubility , Water/chemistry
7.
J Autoimmun ; 37(3): 228-41, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21752599

ABSTRACT

The respective roles of inflammatory and neurodegenerative processes in the pathology of multiple sclerosis (MS) and in its animal model experimental autoimmune encephalomyelitis (EAE) are controversial. Novel treatment strategies aim to operate within the CNS to induce neuroprotection and repair processes in addition to their anti-inflammatory properties. In this study we analyzed and compared the in situ pathological manifestations of EAE utilizing two different models, namely the relapsing-remitting PLP-induced and the chronic MOG-induced diseases. To characterize pathological changes, both transmission electron microscopy (TEM) and immunohistochemistry were employed. The effect of the approved MS drug glatiramer acetate (GA, Copaxone) on myelin damage/repair and on motor neuron loss/preservation was studied in both EAE models. Ultrastructural spinal cord analysis revealed multiple white matter damage foci, with different patterns in the two EAE models. Thus, the relapsing-remitting model was characterized mainly by widespread myelin damage and by remyelinating fibers, whereas in the chronic model axonal degeneration was more prevalent. Loss of lower motor neurons was manifested only in mice with chronic MOG-induced disease. In the GA-treated mice, smaller lesions, increased axonal density and higher prevalence of normal appearing axons were observed, as well as decreased demyelination and degeneration. Furthermore, quantitative analysis of the relative remyelination versus demyelination, provides for the first time evidence of significant augmentation of remyelination after GA treatment. The loss of motor neurons in GA-treated mice was also reduced in comparison to that of EAE untreated mice. These effects were obtained even when GA treatment was applied in a therapeutic schedule, namely after the appearance of clinical symptoms. Hence, the remyelination and neuronal preservation induced by GA are in support of the neuroprotective consequences of this treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/pathology , Multiple Sclerosis, Relapsing-Remitting/pathology , Nerve Degeneration/prevention & control , Neuroprotective Agents/administration & dosage , Peptides/administration & dosage , Spinal Cord/pathology , Animals , Axons/drug effects , Axons/ultrastructure , Chronic Disease , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Glatiramer Acetate , Humans , Immunohistochemistry , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Motor Neurons/drug effects , Motor Neurons/ultrastructure , Multiple Sclerosis, Relapsing-Remitting/chemically induced , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Myelin Proteins/administration & dosage , Myelin Proteins/adverse effects , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/adverse effects , Myelin Sheath/drug effects , Myelin Sheath/ultrastructure , Myelin-Associated Glycoprotein/ultrastructure , Myelin-Oligodendrocyte Glycoprotein , Neuroprotective Agents/therapeutic use , Peptides/therapeutic use , Spinal Cord/drug effects , Spinal Cord/ultrastructure
8.
APMIS ; 119(6): 336-46, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21569091

ABSTRACT

In this study we demonstrate that experimental autoimmune encephalomyelitis (EAE) induced by the MBP-PLP fusion protein MP4, MOG peptide 35-55, or PLP peptide 178-191 in C57BL/6 mice, respectively, displays distinct features of CNS pathology. Major differences between the three models resided in (i) the region-/tract-specificity and disseminated nature of spinal cord degeneration, (ii) the extent and kinetics of demyelination, and (iii) the involvement of motoneurons in the disease. In contrast, axonal damage was present in all models and to a similar extent, proposing this feature as a possible morphological correlate for the comparable chronic clinical course of the disease induced by the three antigens. The data suggest that the antigen targeted in autoimmune encephalomyelitis is crucial to the induction of differential histopathological disease manifestations. The use of MP4-, MOG:35-55-, and PLP:178-191-induced EAE on the C57BL/6 background can be a valuable tool when it comes to reproducing and studying the structural-morphological diversity of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Myelin Proteolipid Protein/immunology , Myelin-Associated Glycoprotein/immunology , Peptide Fragments/immunology , Spinal Cord/pathology , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Mice , Mice, Inbred C57BL , Motor Neuron Disease/pathology , Myelin Basic Protein/immunology , Myelin Proteins , Myelin Proteolipid Protein/adverse effects , Myelin-Associated Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/adverse effects , Phenotype , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/immunology
9.
Ann Neurol ; 69(5): 878-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21391234

ABSTRACT

OBJECTIVE: The subventricular zone (SVZ) of the brain constitutes a niche for neural stem and progenitor cells that can initiate repair after central nervous system (CNS) injury. In a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE), the neural stem cells (NSCs) become activated and initiate regeneration during acute disease, but lose this ability during the chronic phases of disease. We hypothesized that chronic microglia activation contributes to the failure of the NSC repair potential in the SVZ. METHODS: Using bromodeoxyuridine injections at different time points during EAE, we quantified the number of proliferating and differentiating progenitors, and evaluated the structure of the SVZ by electron microscopy. In vivo minocycline treatment during EAE was used to address the effect of microglia inactivation on SVZ dysfunction. RESULTS: In vivo treatment with minocycline, an inhibitor of microglia activation, increases stem cell proliferation in both naive and EAE animals. Minocycline treatment decreases cortical and periventricular pathology in the chronic phase of EAE, improving the proliferation of Sox2 stem cells and NG2 oligodendrocyte precursors cells originating in the SVZ and their differentiation into mature oligodendrocytes. INTERPRETATION: These data suggest that failure of repair observed during chronic EAE correlates with microglia activation and that treatments targeting chronic microglial activation have the potential for enhancing repair in the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Neural Stem Cells/physiology , Stem Cell Niche/pathology , Animals , Anti-Bacterial Agents/pharmacology , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Mice , Microglia/drug effects , Microglia/pathology , Microglia/ultrastructure , Microscopy, Electron, Transmission , Minocycline/pharmacology , Multiple Sclerosis , Myelin Proteolipid Protein/adverse effects , Neural Stem Cells/drug effects , Neural Stem Cells/ultrastructure , Oligodendroglia/drug effects , Oligodendroglia/physiology , Peptide Fragments/adverse effects , Secondary Prevention , Stem Cell Niche/drug effects , Time Factors
10.
J Neurosci ; 30(27): 9074-83, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20610741

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS and remyelination in MS ultimately fails. Although strategies to promote myelin repair are eagerly sought, mechanisms underlying remyelination in vivo have been elusive. CXCR2 is expressed on neutrophils and oligodendrocyte lineage cells in the CNS. CXCR2-positive neutrophils facilitate inflammatory demyelination in demyelination models such as experimental autoimmune encephalomyelitis (EAE) and cuprizone intoxication. Systemic injection of a small molecule CXCR2 antagonist at the onset of EAE decreased demyelinated lesions. These results left the cellular target of the CXCR2 antagonist uncertain and did not clarify whether CXCR2 blockade prevented demyelination or promoted remyelination. Here, we show that the actions of CXCR2 on nonhematopoietic cells unexpectedly delay myelin repair. Bone marrow chimeric mice (Cxcr2(+/-)-->Cxcr2(-/-) and Cxcr2(+/-)-->Cxcr2(+/+)) were subjected to two distinct models of myelin injury. In all cases, myelin repair was more efficient in Cxcr2(+/-)-->Cxcr2(-/-) animals. Oligodendrocyte progenitor cells (OPCs) in demyelinated lesions of Cxcr2(+/-)-->Cxcr2(-/-) mice proliferated earlier and more vigorously than in tissues from Cxcr2(+/-)--> Cxcr2(+/+) animals. In vitro demyelinated CNS slice cultures also showed better myelin repair when CXCR2 was blocked with neutralizing antibodies or was genetically deleted. Our results suggest that CXCR2 inactivation permits optimal spatiotemporal positioning of OPCs in demyelinating lesions to receive local proliferative and differentiating signals. Given that CXCR2 exerts dual functions that promote demyelination and decrease remyelination by actions toward hematopoietic cells and nonhematopoietic cells, respectively, our findings identify CXCR2 as a promising drug target for clinical demyelinating disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Myelin Sheath/physiology , Nerve Regeneration/physiology , Receptors, Interleukin-8B/metabolism , Recovery of Function/physiology , Animals , Animals, Newborn , Antibodies/pharmacology , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Differentiation/drug effects , Central Nervous System Stimulants/pharmacology , Cerebellum/drug effects , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Freund's Adjuvant/adverse effects , Glycoproteins/adverse effects , In Vitro Techniques , Leukocyte Common Antigens/metabolism , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission/methods , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/adverse effects , Myelin Sheath/ultrastructure , Myelin-Oligodendrocyte Glycoprotein , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Neurologic Examination , Oligodendroglia/drug effects , Oligodendroglia/physiology , Peptide Fragments/adverse effects , Picrotoxin/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Recovery of Function/drug effects , Recovery of Function/genetics , Severity of Illness Index , Stem Cells/drug effects , Time Factors
11.
Neurobiol Dis ; 32(2): 273-80, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18778774

ABSTRACT

We investigated the treatment of experimental autoimmune encephalomyelitis (EAE) in mice with Niaspan, an agent used to elevate high-density lipoprotein (HDL). EAE mice were treated with Niaspan starting on the immunization or clinical onset day. Neurological functional recovery was significantly increased in the Niaspan treated mice (100 mg/kgbw) compared to the controls. Inflammatory infiltrates were significantly reduced in the Niaspan treatment group compared to the EAE controls. HDL level, intact myelin area, newly formed oligodendrocytes, regenerating axons, gene and protein levels of sonic hedgehog (Shh)/Gli1 were significantly increased in the Niaspan treated mice compared to EAE controls. These data indicate that Niaspan treatment improved functional recovery after EAE, possibly, via reducing inflammatory infiltrates and demyelination areas, and stimulating oligodendrogenesis and axonal regeneration. Niaspan-mediated activation of Shh/Gli1 pathway may promote functional recovery post-EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Niacin/therapeutic use , Recovery of Function/drug effects , Vitamin B Complex/therapeutic use , Animals , Cell Line, Transformed , Cell Proliferation/drug effects , Cholesterol, HDL/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Myelin Proteolipid Protein/adverse effects , Nerve Regeneration/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Signal Transduction/drug effects , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , Time Factors
12.
Clin Immunol ; 129(1): 69-79, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18676182

ABSTRACT

The objective was to optimize and evaluate the in vivo activities of our novel bifunctional peptide inhibitor (BPI), which alters immune response in autoimmune diseases by modulating the immunological synapse formation. Previously, we have designed PLP-BPI and GAD-BPI by conjugating myelin proteolipid protein (PLP)(139-151) and glutamic acid decarboxylase (GAD)(208-217), respectively, with CD11a(237-246) via a spacer peptide. PLP-BPI and GAD-BPI suppressed the disease progression in experimental autoimmune encephalomyelitis (EAE) and in type-1 diabetes, respectively. In this study, various PLP-BPI derivatives were synthesized and evaluated in the EAE model. Intravenous injections of PLP-BPI derivatives prevented the disease progression more efficiently than did unmodified PLP-BPI. Production of IL-17, a potent proinflammatory cytokine found commonly among MS patients, was significantly low in Ac-PLP-BPI-NH(2)-2-treated mice. Treatment given after the disease onset could dramatically ameliorate the disease. BPI induced anaphylactic responses at a lower incidence than PLP(139-151). In conclusion, PLP-BPI derivatives can effectively suppress the disease severity and morbidity of EAE by post-onset therapeutic treatment as well as prophylactic use.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interleukin-17/blood , Myelin Proteolipid Protein/therapeutic use , Peptide Fragments/therapeutic use , Anaphylaxis/chemically induced , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/immunology , Peptide Fragments/adverse effects , Peptide Fragments/immunology
13.
J Immunol ; 178(8): 4749-56, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404254

ABSTRACT

Administration of autoantigens under conditions that induce type 2 immunity frequently leads to protection from T cell-mediated autoimmune diseases. Such treatments, however, are inherently linked to the induction of IgG1 Abs and to the risk of triggering anaphylactic reactions. We studied the therapeutic benefit vs risk of immune deviation in experimental allergic encephalomyelitis of SJL mice induced by MP4, a myelin basic protein-proteolipid protein (PLP) fusion protein. MP4 administration in IFA induced type 2 T cell immunity, IgG1 Abs, and experimental allergic encephalomyelitis protection, and all three were enhanced by repeat injections. Despite high Ab titers, anaphylactic side reactions were not observed when MP4 was repeatedly injected in IFA or as soluble Ag s.c. In contrast, lethal anaphylaxis was seen after s.c. injection of soluble PLP:139-151 peptide, but not when the peptide was reinjected in IFA. Therefore, the Ab response accompanying the immune therapy constituted an anaphylactic risk factor only when the autoantigen was not retained in an adjuvant and when it was small enough to be readily disseminated within the body. Taken together, our data show that treatment regimens can be designed to boost the protective type 2 T cell response while avoiding the risk of Ab-mediated allergic side effects.


Subject(s)
Anaphylaxis/etiology , Autoantigens/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/therapy , Myelin Basic Protein/therapeutic use , Myelin Proteolipid Protein/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Autoantigens/administration & dosage , Autoantigens/adverse effects , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunization , Immunoglobulin G/biosynthesis , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Mice , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/adverse effects , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/immunology , Nerve Tissue Proteins/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/adverse effects , T-Lymphocytes/immunology
14.
J Immunol ; 178(3): 1372-8, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17237384

ABSTRACT

In experimental autoimmune encephalomyelitis (EAE), the production of proinflammatory cytokines by neuroantigen-specific T cells is thought to initiate and maintain the inflammatory autoimmune pathology. Because gene knockout strategies have shown that IFN-gamma and TNF are not essential for EAE development, there is increasing interest in establishing the role of other proinflammatory cytokines, primarily IL-17 in EAE. We used an IL-17 ELISPOT assay to track the neuroantigen-specific IL-17-producing T cells at single-cell resolution in various organs of SJL mice undergoing PLP 139-151-induced EAE. Overall, the migration patterns and population kinetics of the PLP 139-151-specific IL-17-producing CD4 cells were reminiscent of the IFN-gamma-producing cells, with the exception of IL-17 producers far outnumbering the IFN-gamma and IL-2 producers in the inflamed CNS. The selective enrichment of IL-17-producing CD4 cells in the CNS is suggestive of the pathogenic role of an independent (non-Th1) IL-17-producing proinflammatory effector T cell class in EAE.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/analysis , Animals , Central Nervous System/cytology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Inflammation/etiology , Interferon-gamma , Interleukin-2 , Kinetics , Mice , Mice, Inbred Strains , Myelin Proteolipid Protein/adverse effects , Organ Specificity , Peptide Fragments/adverse effects , T-Lymphocytes, Helper-Inducer
15.
J Immunol ; 169(1): 117-25, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12077236

ABSTRACT

Pertussis toxin (PT) has been widely used to facilitate the induction of experimental autoimmune encephalomyelitis (EAE) in rodents. It has been suggested that this microbial product promotes EAE by opening up the blood-brain barrier and thereby facilitates the migration of pathogenic T cells to the CNS. However, PT has other biological effects that could contribute to its activity in EAE, such as enhancing the cytokine production by T cells and induction of lymphocytosis. In this work, we investigated the effects of PT on the pathogenicity, cytokine differentiation, and clonal sizes of neuroantigen-reactive T cells in EAE in mice. Our results show that PT prevented the protection from EAE conferred by injection of PLPp139-151 in IFA and induced high frequencies of peptide-specific Th1 cells and disease. Interestingly, the mice developed EAE despite the simultaneous vigorous clonal expansion of PLPp139-151-specific Th2 cells. The data indicate that the Th2 cells in this model neither were protective against EAE nor promoted the disease. Furthermore, the results suggested that the effects of the toxin on neuroantigen-reactive T cells were promoted by the PT-induced activation of APCs in lymphoid tissues and the CNS. Together, the results suggest that microbial products, such as PT, could contribute to the initiation of autoimmune disease by modulating the interaction between the innate and adaptive immune system in the response to self Ags.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Freund's Adjuvant/administration & dosage , Lipids , Lymphocyte Activation/immunology , Myelin Basic Protein/immunology , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Pertussis Toxin , Th1 Cells/immunology , Th2 Cells/cytology , Virulence Factors, Bordetella/pharmacology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , Cell Movement/immunology , Clone Cells/cytology , Clone Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Freund's Adjuvant/adverse effects , Freund's Adjuvant/immunology , Freund's Adjuvant/pharmacology , Immune Tolerance , Injections, Intraperitoneal , Injections, Subcutaneous , Lymphocyte Count , Mice , Mice, Inbred Strains , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/therapeutic use , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/therapeutic use , Peptide Fragments/administration & dosage , Peptide Fragments/adverse effects , Peptide Fragments/therapeutic use , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord/pathology , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/transplantation , Th1 Cells/cytology , Th2 Cells/immunology , Th2 Cells/transplantation
16.
Nat Immunol ; 2(7): 605-11, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429544

ABSTRACT

The inducible costimulatory molecule (ICOS) is expressed on activated T cells and participates in a variety of important immunoregulatory functions. After the induction of experimental allergic encephalomyelitis in SJL mice with proteolipid protein (PLP), brain ICOS mRNA and protein were up-regulated on infiltrating CD3+ T cells before disease onset. ICOS blockade during the efferent immune response (9-20 days after immunization) abrogated disease, but blockade during antigen priming (1-10 days after immunization) exacerbated disease. Upon culture with PLP and compared with immunized controls, splenocytes produced either decreased interferon-gamma (IFN-gamma, in efferent blockade) or excessive IFN-gamma (in priming blockade). PLP-specific immunoglobulin G1 was decreased in animals treated with anti-ICOS during antigen priming, but not in other groups.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Brain/immunology , Brain/pathology , Cytokines/biosynthesis , Cytokines/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunoglobulin G/biosynthesis , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Interferon-gamma/biosynthesis , Mice , Myelin Proteolipid Protein/adverse effects , Myelin Proteolipid Protein/immunology , T-Lymphocytes/immunology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...