Science Immunology | 2021

Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals

 
 
 
 
 
 
 
 
 
 
 
 

Abstract


Sustained activation of mTORC1 at late endosomes in primary CD4+ T cell responses of older individuals impairs T helper cell function. Rewiring aged T cells Age-associated decline in T cell function contributes to impaired immune responses to infection and vaccination. Effector versus memory T cell differentiation is controlled in part by signaling and metabolic reprogramming mediated by mechanistic target of rapamycin complex 1 (mTORC1), which is typically activated at amino acid–producing lysosomes. Jin et al. demonstrate that in naïve CD4+ T cells from older individuals, mTORC1 activation instead occurs at late endosomes and depends on the amino acid transporter SLC7A5. Late endosomal mTORC1 impaired T cell lysosomal function, reducing degradation of PD-1 and proliferative responses. Silencing VPS39, a gene that promotes late endosome formation, could increase proliferation of aged human T cells and memory responses of lysosome-defective T cells in LCMV-infected mice, demonstrating that targeting late endosomal mTORC1 activity may improve T cell function. The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.

Volume 6
Pages None
DOI 10.1126/sciimmunol.abg0791
Language English
Journal Science Immunology

Full Text