Activation of mammalian target of rapamycin controls the loss of TCRζ in lupus T cells through HRES-1/Rab4-regulated lysosomal degradation

DR Fernandez, T Telarico, E Bonilla, Q Li… - The Journal of …, 2009 - journals.aai.org
DR Fernandez, T Telarico, E Bonilla, Q Li, S Banerjee, FA Middleton, PE Phillips, MK Crow
The Journal of Immunology, 2009journals.aai.org
Persistent mitochondrial hyperpolarization (MHP) and enhanced calcium fluxing underlie
aberrant T cell activation and death pathway selection in systemic lupus erythematosus.
Treatment with rapamycin, which effectively controls disease activity, normalizes CD3/CD28-
induced calcium fluxing but fails to influence MHP, suggesting that altered calcium fluxing is
downstream or independent of mitochondrial dysfunction. In this article, we show that activity
of the mammalian target of rapamycin (mTOR), which is a sensor of the mitochondrial …
Abstract
Persistent mitochondrial hyperpolarization (MHP) and enhanced calcium fluxing underlie aberrant T cell activation and death pathway selection in systemic lupus erythematosus. Treatment with rapamycin, which effectively controls disease activity, normalizes CD3/CD28-induced calcium fluxing but fails to influence MHP, suggesting that altered calcium fluxing is downstream or independent of mitochondrial dysfunction. In this article, we show that activity of the mammalian target of rapamycin (mTOR), which is a sensor of the mitochondrial transmembrane potential, is increased in lupus T cells. Activation of mTOR was inducible by NO, a key trigger of MHP, which in turn enhanced the expression of HRES-1/Rab4, a small GTPase that regulates recycling of surface receptors through early endosomes. Expression of HRES-1/Rab4 was increased in CD4+ lupus T cells, and in accordance with its dominant impact on the endocytic recycling of CD4, it was inversely correlated with diminished CD4 expression. HRES-1/Rab4 overexpression was also inversely correlated with diminished TCRζ protein levels. Pull-down studies revealed a direct interaction of HRES-1/Rab4 with CD4 and TCRζ. Importantly, the deficiency of the TCRζ chain and of Lck and the compensatory up-regulation of FcεRIγ and Syk, which mediate enhanced calcium fluxing in lupus T cells, were reversed in patients treated with rapamcyin in vivo. Knockdown of HRES-1/Rab4 by small interfering RNA and inhibitors of lysosomal function augmented TCRζ protein levels in vitro. The results suggest that activation of mTOR causes the loss of TCRζ in lupus T cells through HRES-1/Rab4-dependent lysosomal degradation.
journals.aai.org