20 research outputs found

    iNOS-deficiency does not impact polyp number in APC<sup>Min/+</sup>MSH2

    No full text
    <p><sup>βˆ’/βˆ’ </sup><b>mice.</b> Polyp count in the small intestine (A) or in the colon (B) of APC<sup>Min/+</sup>MSH2<sup>+/βˆ’</sup> (A<sup>+/βˆ’</sup>M<sup>+/βˆ’</sup>) mice and APC<sup>Min/+</sup>MSH2<sup>βˆ’/βˆ’</sup> (A<sup>+/βˆ’</sup>M<sup>βˆ’/βˆ’</sup>) mice that were iNOS proficient (N<sup>+/βˆ’</sup>) or iNOS deficient (N<sup>βˆ’/βˆ’</sup>).</p

    Nitric oxide induces DNA mutations that are repaired by the MMR pathway.

    No full text
    <p>(A) Measurements of the amounts of nitric oxide in culture medium generated by various concentrations of SNAP or by macrophages that were stimulated by LPS and IFN-Ξ³. (B) The mutant frequency at the <i>lac</i>I gene in Msh2<sup>+/βˆ’</sup> (WT, nβ€Š=β€Š3) and Msh2<sup>βˆ’/βˆ’</sup> (nβ€Š=β€Š3) SNAP-treated or non-treated macrophages was measured by the Big Blue mutagenesis screen. Bone marrow-derived macrophages were cultured in presence of 150 Β΅M SNAP for 20 hrs. Genomic DNA was isolated and the mutant frequency at <i>lac</i>I gene was calculated. (C) Mutation spectrum in SNAP-treated macrophages as determined by sequencing analysis. The C:G β†’ T:A transitions are shown. The tranversion mutations include C:G β†’ A:T, C:G β†’ G:C, T:A β†’ G:C and T:A β†’ A:T substitutions. The insertions and deletions are presents as other mutations. (D) The mutant frequency at the <i>hprt</i> gene in Msh2<sup>+/βˆ’</sup> (WT, nβ€Š=β€Š8) and Msh2<sup>βˆ’/βˆ’</sup> (nβ€Š=β€Š8) SNAP-treated or non-treated Pre-B cells. (E) The mutant frequency at the <i>hprt</i> gene in SW680 (nβ€Š=β€Š8) and DLD-1 (nβ€Š=β€Š8) SNAP-treated or non-treated human colorectal cancer cell lines.</p

    Anti-MOG antibodies generated in WT and AID<sup>βˆ’/βˆ’</sup> mice.

    No full text
    <p>WT and AID<sup>βˆ’/βˆ’</sup> mice were immunized with recombinant human MOG (rhMOG) and titres of anti-MOG Ab were evaluated at day 15 (peak of disease). As expected, AID<sup>βˆ’/βˆ’</sup> mice only made anti-MOG Ab of the IgM isotype, whereas WT mice produced anti-MOG IgG1 and IgG2c Ab (and very little IgM at this time point). The average OD for each mouse at a set concentration of serum (half-way point of the curve) is depicted, and raw OD values can be seen in Supplemental <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0061478#pone.0061478.s003" target="_blank">Figure S3</a>. In this experiment, 5–6 mice per group were tested and an additional experiment tested 8–12 mice.</p

    Secondary B Cell Receptor Diversification Is Necessary for T Cell Mediated Neuro-Inflammation during Experimental Autoimmune Encephalomyelitis

    Get PDF
    <div><p>Background</p><p>Clinical studies of B cell depletion in Multiple Sclerosis (MS) have revealed that B Lymphocytes are involved in the neuro-inflammatory process, yet it remains unclear how B cells can exert pro- and anti-inflammatory functions during MS. Experimental Autoimmune Encephalomyelitis (EAE) is an animal model of MS whereby myelin-specific T cells become activated and subsequently migrate to the Central Nervous System (CNS) where they perform pro-inflammatory functions such as cytokine secretion. Typically EAE is induced by immunization of mice of a susceptible genetic background with peptide antigen emulsified in Complete Freund's Adjuvant. However, novel roles for B-lymphocytes in EAE may also be explored by immunization with full-length myelin oligodendrocyte glycoprotein (MOG) that contains the B cell conformational epitope. Here we show that full length MOG immunization promotes a chronic disease in mice that depends on antigen-driven secondary diversification of the B cell receptor.</p><p>Methods</p><p>Activation-Induced Deaminase (AID) is an enzyme that is essential for antigen-driven secondary diversification of the B cell receptor. We immunized AID<sup>βˆ’/βˆ’</sup> mice with the extracellular domain (amino acids 1–120) of recombinant human MOG protein (rhMOG) and examined the incidence and severity of disease in AID<sup>βˆ’/βˆ’</sup> versus wild type mice. Corresponding with these clinical measurements, we also evaluated parameters of T cell activation in the periphery and the CNS as well as the generation of anti-MOG antibodies (Ab).</p><p>Conclusions</p><p>AID<sup>βˆ’/βˆ’</sup> mice exhibit reduced severity and incidence of EAE. This suggests that the secondary diversification of the B cell receptor is required for B cells to exert their full encephalogenic potential during rhMOG-induced EAE, and possibly also during MS.</p></div

    Appearance of class switched B cells in the CNS during EAE.

    No full text
    <p>WT and AID<sup>βˆ’/βˆ’</sup> mice were immunized with recombinant human MOG (rhMOG) and spinal cords were extracted and processed at day 15 (peak of disease). B cells were identified by the expression of CD19 and B220. Class switched B cells were further identified by the absence of IgM/IgD and reported here as a percentage of the total number of B220<sup>+</sup>CD19<sup>+</sup> B cells. Representative FACS analysis can be seen in Supplemental <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0061478#pone.0061478.s002" target="_blank">Figure S2B</a>.</p

    Lymph node cells from WT and AID<sup>βˆ’/βˆ’</sup> mice produce equivalent levels of cytokines in response to immunization with rhMOG.

    No full text
    <p>WT and AID<sup>βˆ’/βˆ’</sup> mice were immunized with recombinant human MOG (rhMOG) and draining axillary and brachial lymph nodes were harvested after 7 days post-immunization. Four million lymph node cells were plated along with 20 Β΅g of rhMOG. Cultures were kept for 48 hours after which the supernatant was harvested and evaluated for cytokine production by ELISA. The limit of detection of the assay is 125 pg/ml for the IFNΞ³ ELISA and 63 pg/ml for the IL-17 ELISA. Five mice per group were assessed.</p

    Characteristics of the mutations within the <i>lacI</i> gene in Msh2 WT and Msh2<sup>βˆ’/βˆ’</sup> macrophages that were SNAP-treated or untreated.

    No full text
    <p>Characteristics of the mutations within the <i>lacI</i> gene in Msh2 WT and Msh2<sup>βˆ’/βˆ’</sup> macrophages that were SNAP-treated or untreated.</p

    Impaired accumulation of cytokine producing CD4<sup>+</sup> T cells in the CNS of AID<sup>βˆ’/βˆ’</sup> mice.

    No full text
    <p>WT and AID<sup>βˆ’/βˆ’</sup> mice were immunized with recombinant human MOG (rhMOG) and spinal cords (<b>A</b>) and brains (<b>B</b>) were extracted and processed at day 15: the peak of disease. Leukocytes were stimulated <i>ex vivo</i> with PMA/Ionomycin, and brefeldin A was added in the last 4 hours. Cells were then subjected to surface and intracellular cytokine staining. This experiment was performed twice with similar results and 6 mice per group were tested in this experiment. Representative FACS analysis can be seen in Supplemental <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0061478#pone.0061478.s002" target="_blank">Figure S2A</a>.</p

    AID<sup>βˆ’/βˆ’</sup> mice exhibit attenuated EAE in response to rhMOG but not MOG<sub>35-55</sub>.

    No full text
    <p>(<b>A</b>) WT and AID<sup>βˆ’/βˆ’</sup> mice were immunized with MOG<sub>35-55</sub> in CFA and examined for clinical symptoms. A representative experiment is shown, and 2 experiments were performed with similar results (shown here are nβ€Š=β€Š6 WT and nβ€Š=β€Š5 AID<sup>βˆ’/βˆ’</sup> mice per group). (<b>B</b>) WT and AID<sup>βˆ’/βˆ’</sup> mice were immunized with rhMOG in CFA and examined for clinical symptoms. A representative experiment is shown (see <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0061478#pone-0061478-t001" target="_blank">Table 1</a> for all 4 experiments) with nβ€Š=β€Š11 WT and nβ€Š=β€Š7 AID<sup>βˆ’/βˆ’</sup> mice per group. At the termination of the experiment, spinal cords were dissected and stained with H&E and counterstained with Luxol fast blue. A representative example of 6 separate mice is shown (original magnification Γ—200). See arrows for areas of cellular infiltrates (<b>C</b>). Cell infiltration in the spinal cord tissue was assessed as follows: meningeal infiltrate (score 1), perivascular infiltrate (score 2), parenchymal infiltrate (score 3) (<b>D</b>).</p
    corecore