Credit: PHOTODISC

The complement and Notch systems have well-appreciated roles in innate immunity and immune cell development, respectively. A recent study in Nature Immunology suggests that crosstalk between these two systems also regulates adaptive T cell responses in humans.

a role for jagged–CD46 interactions in regulating human TH1 cell responses

CD46 (also known as membrane cofactor protein) protects host cells from complement-mediated damage by binding to and promoting the inactivation of complement components C3b and C4b. There is also evidence that CD46 regulates the induction of T helper 1 (TH1) cells in humans and their subsequent switching to an interferon-γ (IFNγ)+ and interleukin-10 (IL-10)+ phenotype and then to an IL-10+ regulatory phenotype. The authors suspected that an alternative ligand for CD46 may exist, which could explain these complex effects on TH1 cell responses. Indeed, initial screens identified the Notch ligand jagged 1 as a ligand for CD46.

They next explored whether CD46–jagged 1 interactions influence T cell responses. Activation of human T cells with CD3- and CD46-specific antibodies in vitro led to their upregulation of Notch 1, Notch 2, jagged 1 and jagged 2, but caused the downregulation of the Notch ligand Delta-like 1 (DLL1) and of CD46 itself. The upregulation of these Notch proteins seems to be crucial for CD46-mediated control of TH1 cell responses, as blocking Notch signalling pathways prevented CD46 from driving the upregulation of IFNγ by human TH1 cells and their subsequent switching to an IFNγ+IL-10+ phenotype. Confocal microscopy studies showed that jagged 1 mainly colocalized with CD46 and not with Notch in resting T cells. However, following CD3- and CD46-mediated activation of T cells, 50% of Notch became associated with jagged 1. This suggests that the sequential engagement of CD46 and then Notch by jagged 1 regulates the upregulation of IFNγ by TH1 cells and subsequent switching to dual IFNγ and IL-10 production.

To further investigate the importance of this pathway, the authors examined T cells from three patients with mutations in CD46. Notably, these patients suffer from recurrent infections. Each patient had normal numbers of B cells, CD4+ T cells and CD8+ T cells. Furthermore, following stimulation in vitro, their T cells proliferated normally and differentiated into TH2-type effectors. However, T cells from two of the patients were unable to differentiate into TH1-type effector cells. In addition, although T cells from the third patient could differentiate into TH1-type effectors, they were unable to switch to an IFNγ+IL-10+ regulatory phenotype.

Finally, the authors assessed T cells from four patients with Alagille's syndrome. These individuals have heterozygous mutations in the gene encoding jagged 1 that cause developmental defects. Some patients also suffer from recurrent infections, but the reasons for this have not been understood. Similarly to the T cells from patients with CD46 mutations, T cells from the patients with Alagille's syndrome showed defective IFNγ induction and were unable to switch to an IFNγ+IL-10+ phenotype. Interestingly, activated T cells from patients with Alagille's syndrome or with CD46 mutations had deregulated expression of CD127 and CD132; the authors propose that jagged–CD46 interactions may influence T cell signalling in response to IL-2 family members. Taken together, these data describe a role for jagged–CD46 interactions in regulating human TH1 cell responses and suggest that the failure of this pathway can increase susceptibility to infection.