15 research outputs found

    Slamf6 negatively regulates autoimmunity

    Get PDF
    The nine SLAM family (Slamf) receptors are positive or negative regulators of adaptive and innate immune responses, and of several autoimmune diseases. Here we report that the transfer of Slamf6-/- B6 CD4+ T cells into co-isogenic bm12 mice causes SLE-like autoimmunity with elevated levels of autoantibodies. In addition, significantly higher percentages of Tfh cells and IFN-γ-producing CD4+ cells, as well as GC B cells were observed. Interestingly, the expression of the Slamf6-H1 isoform in Slamf6-/- CD4+ T cells did not induce this lupus-like phenotype. By contrast, Slamf1-/- or Slamf5-/- CD4+ T cells caused the same pathology as WT CD4+ T cells. As the transfer of Slamf [1+6]-/- or Slamf [1+5+6]-/- CD4+ T cells induced WT levels of autoantibodies, the presence of Slamf1 was requisite for the induction of increased levels of autoantibodies by Slamf6-/- CD4+ T cells. We conclude that Slamf6 functions as an inhibitory receptor that controls autoimmune responses

    A combination of an anti-SLAMF6 antibody and ibrutinib efficiently abrogates expansion of chronic lymphocytic leukemia cells

    Get PDF
    The signaling lymphocyte activation molecule family [SLAMF] of cell surface receptors partakes in both the development of several immunocyte lineages and innate and adaptive immune responses in humans and mice. For instance, the homophilic molecule SLAMF6 (CD352) is in part involved in natural killer T cell development, but also modulates T follicular helper cell and germinal B cell interactions. Here we report that upon transplantation of a well-defined aggressive murine B220+CD5+ Chronic Lymphocytic Leukemia (CLL) cell clone, TCL1-192, into SCID mice one injection of a monoclonal antibody directed against SLAMF6 (αSlamf6) abrogates tumor progression in the spleen, bone marrow and blood. Similarly, progression of a murine B cell lymphoma, LMP2A/λMyc, was also eliminated by αSlamf6. But, surprisingly, αSLAMF6 neither eliminated TCL1-192 nor LMP2A/λMyc cells, which resided in the peritoneal cavity or omentum. This appeared to be dependent upon the tumor environment, which affected the frequency of sub-populations of the TCL1-192 clone or the inability of peritoneal macrophages to induce Antibody Dependent Cellular Cytotoxicity (ADCC). However, co-administering αSlamf6 with the Bruton tyrosine kinase (Btk) inhibitor, ibrutinib, synergized to efficiently eliminate the tumor cells in the spleen, bone marrow, liver and the peritoneal cavity. Because an anti-human SLAMF6 mAb efficiently killed human CLL cells in vitro and in vivo, we propose that a combination of αSlamf6 with ibrutinib should be considered as a novel therapeutic approach for CLL and other B cell tumors

    Slamf6 negatively regulates autoimmunity

    No full text
    The nine SLAM family (Slamf) receptors are positive or negative regulators of adaptive and innate immune responses, and of several autoimmune diseases. Here we report that the transfer of Slamf6-/- B6 CD4+ T cells into co-isogenic bm12 mice causes SLE-like autoimmunity with elevated levels of autoantibodies. In addition, significantly higher percentages of Tfh cells and IFN-γ-producing CD4+ cells, as well as GC B cells were observed. Interestingly, the expression of the Slamf6-H1 isoform in Slamf6-/- CD4+ T cells did not induce this lupus-like phenotype. By contrast, Slamf1-/- or Slamf5-/- CD4+ T cells caused the same pathology as WT CD4+ T cells. As the transfer of Slamf [1+6]-/- or Slamf [1+5+6]-/- CD4+ T cells induced WT levels of autoantibodies, the presence of Slamf1 was requisite for the induction of increased levels of autoantibodies by Slamf6-/- CD4+ T cells. We conclude that Slamf6 functions as an inhibitory receptor that controls autoimmune responses

    A combination of an anti-SLAMF6 antibody and ibrutinib efficiently abrogates expansion of chronic lymphocytic leukemia cells

    No full text
    The signaling lymphocyte activation molecule family [SLAMF] of cell surface receptors partakes in both the development of several immunocyte lineages and innate and adaptive immune responses in humans and mice. For instance, the homophilic molecule SLAMF6 (CD352) is in part involved in natural killer T cell development, but also modulates T follicular helper cell and germinal B cell interactions. Here we report that upon transplantation of a well-defined aggressive murine B220+CD5+ Chronic Lymphocytic Leukemia (CLL) cell clone, TCL1-192, into SCID mice one injection of a monoclonal antibody directed against SLAMF6 (αSlamf6) abrogates tumor progression in the spleen, bone marrow and blood. Similarly, progression of a murine B cell lymphoma, LMP2A/λMyc, was also eliminated by αSlamf6. But, surprisingly, αSLAMF6 neither eliminated TCL1-192 nor LMP2A/λMyc cells, which resided in the peritoneal cavity or omentum. This appeared to be dependent upon the tumor environment, which affected the frequency of sub-populations of the TCL1-192 clone or the inability of peritoneal macrophages to induce Antibody Dependent Cellular Cytotoxicity (ADCC). However, co-administering αSlamf6 with the Bruton tyrosine kinase (Btk) inhibitor, ibrutinib, synergized to efficiently eliminate the tumor cells in the spleen, bone marrow, liver and the peritoneal cavity. Because an anti-human SLAMF6 mAb efficiently killed human CLL cells in vitro and in vivo, we propose that a combination of αSlamf6 with ibrutinib should be considered as a novel therapeutic approach for CLL and other B cell tumors

    Slamf8 negatively regulates <i>in vitro</i> migration of DCs and of macrophages, while Slamf1 is a positive regulator of the same process.

    No full text
    <p>Wt, Slamf1<sup>-/-</sup>, and Slamf8<sup>-/-</sup> skin DCs were isolated and column-purified before they were allowed to migrate toward a concentration range of CCL21 [0–400nM]. The migration index of <b>(A)</b> Slamf8<sup>-/-</sup> vs. wt DCs and <b>(B)</b> wt vs. Slamf1<sup>-/-</sup> DCs are plotted. <b>(C)</b> Macrophages from wt mice were incubated with different concentrations of DPI (1μM and 5μM) for 15 minutes, and the Nox2 activity in macrophages was quantified upon PMA (1mg/mL) stimulation. The data are representative of two independent experiments. <b>(D)</b> Wt, Slamf1<sup>-/-</sup>, and Slamf8<sup>-/-</sup> thio-macrophages migration in the presence of CSF-1, with and without pre-incubation with DPI. All the data are representative of three independent experiments.</p

    Slamf8<sup>-/-</sup> neutrophil produce higher ROS, and Slamf8 negatively regulates neutrophil migration.

    No full text
    <p><b>(A)</b> ROS production as measured by luminescence in wt and Slamf8<sup>-/-</sup> mice thio-neutrophils in response to heat inactivated <i>E</i>.<i>coli</i> F18 and PMA. <b>(B)</b> Total number of peritoneal cells obtained from wt and Slamf8<sup>-/-</sup> mice after 4 hours <i>i</i>.<i>p</i>. injection of 4% thioglycollate. <b>(C)</b> Number and (<b>D</b>) percentage of neutrophil (CD11b<sup>+</sup> Ly6G<sup>+</sup>) among peritoneal cells in wt and Slamf8<sup>-/-</sup> mice. <b>(E)</b> Representative dot plots (gated on DAPI<sup>-</sup> cells) showing CD11b<sup>+</sup> Ly6G<sup>+</sup> neutrophil in the peritoneal cavity of wt and Slamf8<sup>-/-</sup> mice. <b>(F</b>) Slamf8<sup>-/-</sup> and wt mice were injected with αCD3. Percentage of neutrophils (CD11b<sup>+</sup> Ly6G<sup>high</sup>) from the small intestine lamina propria 0, 3 and 5 days after αCD3 injection. <b>(G)</b> Representative dot plots (gated on CD45.2<sup>+</sup>) showing neutrophils. The data are representative of 2 independent experiments, each consisting of at least 3 mice per experimental condition.</p
    corecore