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 acidтАУproducing lysosomes. Jin et al. demonstrate that in na├пve 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.
Abstract
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.
INTRODUCTION
Aging is associated with a decline in adaptive immunity, resulting in decreased efficacy of vaccination and increased morbidity from infections with newly and previously encountered pathogens (1, 2). Most noticeable are the increased susceptibility during the annual influenza epidemic (3) and, most recently, the increased risk for developing severe disease after SARS-CoV-2 infection (4тАУ6). Although the increased morbidity is clearly multifactorial, age-related changes in CD4+ T cell survival and differentiation have been identified that contribute to the immune decline in older individuals (7, 8). CD4+ T helper cells are pivotal for mounting a protective immune response after vaccination or infections (9). Generation of CD4+ T follicular helper (TFH) cells is essential for germinal center (GC) formation, class switching, and affinity maturation (10). Moreover, CD4+ T cells are required for effective CD8+ T cell memory and recall responses (11тАУ13). Recent studies have shown a T cellтАУintrinsic bias toward short-lived effector cells differentiation in CD4+ T cell responses of older adults at the expense of TFH and memory precursor cells (8). One important mechanism causing this bias is a sustained activation of the mechanistic target of rapamycin complex 1 (mTORC1) (8).
mTORC1 plays an important role in regulating T cell responses by coordinating cell growth and cellular metabolism with environmental inputs, particularly nutrient resources, and facilitating the switch toward anabolic metabolism that is required for cell proliferation and effector cell differentiation (14тАУ16). Complete block of mTORC1 activities by either a treatment with high dose of rapamycin or a genetic deletion of Ras homolog enriched in brain (RHEB) (an upstream activator of mTORC1) markedly reduced the number of antigen-specific effector and memory precursor T cells at peak responses after primary infection (17, 18). RHEB-deficient memory CD8+ T cells even failed to respond to secondary immunization (18). However, overactivation of mTORC1 by genetic depletion of TSC1 (an upstream inhibitor of mTORC1) reduced the numbers of memory precursor CD8+ T cells at peak responses after primary infection instead of promoting them (19тАУ21). Moderate inhibition of mTORC1 by pharmacological compounds produced increased clonal expansion of antigen-specific T cells after primary responses and enhanced memory recall responses after antigen rechallenge in several infection models (22тАУ24). These data led to the conclusion that mTORC1 inhibition promotes long-term memory over short-lived effector CD8+ T cell differentiation (17). Together, fine-tuning of mTORC1 activity is important for generating protective primary and recall T cell responses, particularly for older adults who have sustained mTORC1 activity after T cell activation, resulting in the preferential generation of short-lived effector T cells (8).
mTORC1 translocates to the lysosomal membrane, where it is activated in response to amino acid signaling. In turn, mTORC1 suppresses lysosomal activity by phosphorylating transcription factor EB (TFEB) (25, 26). We previously found that T cells from older adults had lower lysosomal gene expression and proteolytic activities due to reduced TFEB transcription (27). In parallel, mTORC1 activity was more sustained in activated T cells from older adults (8). How lysosome-deficient aged T cells maintain mTORC1 activity remains unresolved. The late endosome is an alternative platform for mTORC1 activation (28тАУ30). Compared with the lysosome, late endosomes have no proteolytic activity and therefore no amino acid efflux (31). They originate by homotypic fusion and vacuole protein sorting (HOPS) complex-mediated conversion from early endosomes (32). Inhibiting early to late endosomal conversion by silencing Vam6/Vps39-like protein (VPS39, a key member of the HOPS complex) led to the formation of hybrid endosomal compartments and attenuated mTORC1 activity (29, 33). Conversely, inhibition of lysosomal activities induced an intracellular expansion of late endosomes (34). Consistent with this observation, an increased number of late endosomes is seen in responses of T cells from older individuals that fail to replenish their lysosomes (27).
Here, we describe that in activated CD4+ T cells from older adults, mTORC1 activation relies on late endosomes rather than lysosomes as an activation platform and the plasma membrane leucine transporter SLC7A5 as an amino acid source. SLC7A5 and VPS39 deficiency reduced mTORC1 activities in T cells from older but not young adults. Sustained activation of late endosomal mTORC1 suppressed lysosomal biogenesis by phosphorylating TFEB, thereby preventing lysosomal degradation of programmed cell death protein-1 (PD-1) and impairing T cell expansion. In vivo, inhibition of late endosome formation by silencing Vps39 in lymphocytic choriomeningitis virus (LCMV)тАУspecific CD4+ SMARTA cells enhanced T cell expansion and improved memory cell transition after LCMV infection, leading to enhanced CD4+ T cell helper responses. Thus, abrogation of late endosomal mTORC1 may be a promising strategy to boost immunity in older individuals.
RESULTS
Lysosome-independent activation of mTORC1 in na├пve CD4+ T cell responses
mTORC1 is recruited to lysosomal membranes where it is activated by released amino acids. However, we found that after 5 days of in vitro stimulation with anti-CD3/anti-CD28 beads, na├пve CD4+ T cells from older (65 to 85 years) adults had impaired lysosomal activity in parallel to enhanced mTORC1 activity, as shown by reduced fluorescence of cleaved dequenched bovine serum albumin (DQ-BSA) and increased S6K1 protein phosphorylation, the downstream effector of mTORC1 activity (Fig. 1A and fig. S1, A and B). To examine this apparent conundrum, we investigated the effects of lysosome inhibition on mTORC1 activity; chloroquine (CQ) and Bafilomycin A1, two lysosomal acidification inhibitors, were used for pharmacological inhibition at nontoxic doses (fig. S1C); TFEB silencing was used as a genetic intervention. All interventions reduced lysosomal activities but unexpectedly neither reduced S6K1 (Fig. 1, B and C, and fig. S1, D to F) nor AKT phosphorylation (fig. S1G), the downstream effector of the growth factor signaling arm of mTORC1 pathway (35).
(A) Na├пve CD4+ T cells were activated with anti-CD3/anti-CD28 beads for 5 days followed by DQ-BSA treatment for 6 hours. Fluorescence of cleaved DQ-BSA was analyzed by flow cytometry to determine lysosomal activities (left). Phospho-S6K1 (Thr389) protein expression was determined by Western blotting (right). Results are from 10 young (Y, 20 to 35 years) and 10 older (O, 65 to 85 years) healthy individuals. Intensities of p-S6K1 protein expression were normalized to ╬▓-actin and are shown relative to the mean from young individuals. MFI, mean fluorescence intensity. (B) Na├пve CD4+ T cells from young individuals were activated for 5 days with the last 2 days in the presence of vehicle, CQ, or bafilomycin A1 (BafA1). Lysosomal activities (left) and mTORC1 activities (right) were determined as in (A). (C) Na├пve CD4+ T cells from young individuals were transfected with control or TFEB siRNA and activated for 5 days. Lysosomal activities (left) and mTORC1 activities (right) were determined. (D) Heat map showing expression differences of genes involved in the amino acid signaling arm of the mTORC1 pathway, comparing the transcriptome of day 5тАУactivated na├пve CD4+ T cells from older and young adults [reanalyzed from (36)]. (E) SLC7A5 and SLC7A1 transcripts from day 5тАУactivated na├пve CD4+ T cells from 12 20- to 35-year-old and 12 65- to 85-year-old healthy adults. Results are expressed relative to the mean from young individuals. (F) Chromatin accessibility at SLC7A5 gene in human EBV, VZV, and influenza (Flu) virusтАУspecific CD4+ T cells from young and older adults. Averaged tracks (young n = 3; older n = 4) at SLC7A5 show increased peak in highlighted (red) region. (G) SLC7A5 and SLC7A1 transcripts in samples from (B) and (C). (H) SLC7A5 and c-MYC protein expression in samples from (B) and (C). (I) c-MYC protein expression in samples from (A). (J) GSEA comparing fold transcript differences in young compared with older na├пve CD4+ T cells on day 5 after stimulation [reanalyzed from (36)] with that of тАЬHallmark_MYC_targetтАЭ. FDR, false discovery rate; NES, normalized enrichment score. (K) SLC7A5 and c-MYC protein expression in day 5тАУstimulated na├пve CD4+ T cells from three older individuals transfected with control or MYC siRNA. Statistical significance by two-tailed unpaired t test (A, E, and I) or two-tailed paired t test (B, C, G, H, and K). *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. NS, not significant;
We then set out to investigate the mechanism of how mTORC1 activity is increased even under the lysosome-deficient conditions in T cell responses of older adults. Because lysosome deficiency likely affects lysosomal efflux of amino acids, we examined expression of genes encoding the amino acid signaling arm of the mTORC1 pathway using data from our recent RNA sequencing (RNA-seq) study comparing na├пve CD4+ T cell responses from young and older adults after activation (36). SLC7A5 that encodes a plasma membrane leucine transporter is selectively more expressed in T cells from older adults, whereas other genes involved in the nutrient arm do not change with age (Fig. 1D). Increased SLC7A5 transcription was confirmed in an independent cohort of day 5тАУactivated na├пve CD4+ T cells from 12 young and 12 older individuals. In contrast, no difference was seen for SLC7A1 that encodes the arginine transporter (Fig. 1E). SLC7A5 expression increased with activation in na├пve CD4+ T cells from young and older donors, but expression was more sustained beyond day 3 in older na├пve CD4+ T cells (fig. S2, A and B). This difference persisted and was still detected for memory CD4+ T cells (fig. S2C). Accordingly, assay for transposase-accessible chromatin using sequencing (ATAC-seq) studies showed increased chromatin accessibility of SLC7A5 in older memory CD4+ T cells in three virus-specific [Epstein-Barr virus (EBV), varicella-zoster virus (VZV), and influenza] systems compared with young cells (Fig. 1F). Consistent with increased SLC7A5 expression in lysosome-deficient aged T cells, pharmacological inhibition of lysosome activity or genetic silencing of TFEB up-regulated SLC7A5 transcript and protein expression (Fig. 1, G and H, and fig. S1H). These data suggested that the up-regulation of SLC7A5 transcription in day 5тАУstimulated na├пve CD4+ T cells from older adults was mechanistically related to their reduced lysosome activity.
Upstream of the differentially opened chromatin sites of SLC7A5 are two c-MYC binding motifs (Fig. 1F). c-MYC is the major driver of SLC7A5 transcription in T cell responses (37). c-MYC protein expression was increased after lysosomal inhibition or TFEB silencing (Fig. 1H), indicating a lysosome-dependent degradation of c-MYC protein. Consistently, day 5тАУactivated aged na├пve CD4+ T cells had higher c-MYC protein level compared with young cells (Fig. 1I). Global gene expression profiles obtained by RNA-seq supported the notion that day 5тАУactivated older na├пve CD4+ T cells had higher c-MYC activity. Gene set enrichment analysis (GSEA) showed that age-associated transcriptional signatures of day 5тАУactivated na├пve CD4+ T cells were correlated with the expression pattern of genes in the Hallmark_MYC_target pathway (Fig. 1J). To confirm that the increased SLC7A5 expression is related to higher c-MYC protein expression, we performed c-MYC silencing in na├пve CD4+ T cells from older adults and found reduced SLC7A5 protein expression (Fig. 1K). Together, these data showed that lysosome deficiency stabilized c-MYC protein, thereby increasing SLC7A5 expression in day 5тАУstimulated na├пve CD4+ T cells from older adults.
SLC7A5-dependent late endosomal mTORC1 activation in na├пve CD4+ T cell responses
To determine whether SLC7A5 activity accounts for the sustained mTORC1 activity, we treated T cell cultures from young and older adults with the specific SLC7A5 inhibitor JPH203. mTORC1 activities were monitored by Western blotting of S6K1 phosphorylation or by flow cytometry of S6RP phosphorylation on days 3 and 5 after activation. In titration experiments, a dose of 5 ╬╝M JPH203 was not cytotoxic (fig. S1C) and inhibited mTORC1 activity by about 50% (fig. S2D). On day 3, SLC7A5 inhibition reduced mTORC1 activity in cells from both young and older adults (Fig. 2, A and B). On day 5, mTORC1 activity was already attenuated in T cell responses of young adults. In contrast, mTORC1 activity remained high in older activated T cells; the excess activity was sensitive to SLC7A5 inhibition (Fig. 2, A and B). To exclude off-target effects of the inhibitor, we performed genetic SLC7A5 silencing and overexpression. Data observed were consistent with those obtained by JPH203 treatment (fig. S2, E to H). Together, activated CD4+ T cells from older adults likely sustained uptake of extracellular amino acids through SLC7A5 to maintain mTORC1 activity despite reduced lysosomal function.
(A and B) Na├пve CD4+ T cells from young and older healthy individuals were activated with anti-CD3/anti-CD28 beads for 3 days in the presence of vehicle or SLC7A5 inhibitor JPH203 (top). Alternatively, cells were activated with anti-CD3/anti-CD28 beads for 5 days with the last 2 days in the presence of vehicle or JPH203 (bottom). mTORC1 activities were determined either by Western blotting of p-S6K1 (A) or by flow cytometry of intracellular p-S6RP (S235/S236) (B). Data are shown as one representative experiment (left) and cumulative data of three or four experiments (right). (C) Na├пve CD4+ T cells were activated for 3 days followed by treatment or not with an AKT inhibitor for 2 hours before harvesting. Endosomes were isolated and analyzed for in vitro mTORC1 kinase activity toward S6K1. Total cell lysates (T) and endosome isolates (E) were analyzed by immunoblotting for indicated proteins. Data are shown as one representative of three experiments. (D) In vitro mTORC1 kinase activity of endosome isolates in day 5тАУstimulated na├пve CD4+ T cells from one young and one older individual. Data are shown as one representative of three experiments. (E) Cells were stained with anti-EEA1, anti-LAMP1, and anti-mTOR. Confocal images representative of two independent experiments are shown. Scale bars, 5 ╬╝m. (F and G) mTORC1 activities in day 3тАУ and day 5тАУstimulated na├пve CD4+ T cells from young and older individuals after control or VPS39 silencing. The gray histogram represents isotype control. Comparison by two-tailed paired t test (A, B, F, and G). *P < 0.05 and **P < 0.01.
We recently described that activated T cells from older adults failed to rebuild their lysosomes and expand the late endosomal compartments (27). To investigate whether these late endosomes provide an alternative platform for mTORC1 signaling in T cells, we isolated endosome fractions from na├пve CD4+ T cells on day 3 after stimulation when mTORC1 activity peaks in both young and older individuals. The endosome isolates were enriched for early and late endosome markers (EEA1, RAB7, and CD63) and depleted of the lysosome markers cathepsin D and cathepsin H, and of the plasma membrane marker sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) pump, indicating high purity of the endosome extracts (Fig. 2C). These endosome isolates contained mTOR, regulatory-associated protein of mTOR (Raptor), and RHEB that are essential for substrate recruitment and kinase activity of mTORC1 (Fig. 2C). In an in vitro kinase assay, these endosome isolates exhibited mTORC1 kinase activity toward the substrate S6K1 that was not seen in endosomes extracted from cells treated with an AKT inhibitor (Fig. 2C). Day 5тАУstimulated na├пve CD4+ T cells from older individuals had reduced lysosomal cathepsins, similar early endosome contents (EEA1), and increased late endosomal compartments, with increased late endosomal mTOR, Raptor, and RHEB protein levels and higher mTORC1 kinase activity as shown by increased S6K1 phosphorylation in the in vitro kinase assay (Fig. 2D). Together, these data indicate that mTORC1 is activated on late endosomes and that late endosomal compartments are expanded in stimulated na├пve CD4+ T cells from older adults accounting for the sustained mTORC1 activity.
We then performed VPS39 silencing to examine whether inhibition of late endosome maturation prevents mTORC1 activation. Confocal imaging confirmed that VPS39 silencing inhibited late endosome maturation and produced hybrid endosomal compartments that contained both the early endosome marker EEA1 and the late endosome/lysosome marker LAMP1 (Fig. 2E). These hybrid compartments colocalized with mTOR (Fig. 2E); however, S6K1 and S6RP phosphorylation were reduced after VPS39 silencing, documenting that these hybrid structures do not support mTORC1 activation (Fig. 2, F and G). T cells from young and older adults exhibited reduced mTORC1 activities after VPS39 silencing on day 3 after T cell stimulation. On day 5, an effect of silencing was only observed in T cells from older adults (Fig. 2, F and G), suggesting that the sustained mTORC1 signaling observed with age depended on the formation of late endosomes. VPS39 has also been implicated in regulating transforming growth factorтАУ╬▓ (TGF-╬▓)/SMAD signaling (38); however, we did not observe changes of SMAD2/3 phosphorylation or reporter activities of a SMAD reporter in T cells after VPS39 silencing (fig. S3, A to C), supporting the notion that the observed reduced mTORC1 activity is due to its impact on endosome maturation and not SMAD signaling.
Sustained activation of late endosomal mTORC1 suppresses lysosomal activities in na├пve CD4+ T cell responses
Our data so far demonstrated that CD4+ T cells from older adults sustained mTORC1 activity on late endosomes that relied on the presence of SLC7A5 and VPS39. mTORC1 phosphorylates TFEB, thereby preventing its transcriptional activity and inhibiting lysosome function (25). mTORC1 inhibition down-regulated TFEB phosphorylation while up-regulating TFEB mRNA and protein levels due to reduced AKT-mediated Forkhead box protein O1 (FOXO1 protein) degradation (fig. S4, A to C). Consistent with this notion, mTORC1 inhibition by Torin 1 partially restored the TFEB-dependent transcription of lysosomal cathepsin genes (CTSB, CTSD, CTSH, and CTSS) (27) and lysosomal proteolytic activities in older, but not young, day 5тАУactivated T cells (Fig. 3, A and B). Inhibition of late endosomal mTORC1 in activated T cells from older adults by either VPS39 silencing or SLC7A5 inhibition had similar effects, while the already higher lysosomal activity in T cells from young adults could not be further increased (Fig. 3, C to E). However, SLC7A5 inhibition could counteract the effects of lysosome inhibition in young T cells. Lysosomal cathepsins and lysosomal activities were increased in CQ and JPH203-cotreated young cells compared with young T cells treated with CQ alone (Fig. 3F). Together, these data demonstrate that sustained activation of mTORC1 in late endosomes suppressed lysosomal activities in day 5тАУstimulated na├пve CD4+ T cells from older adults.
(A to F) Na├пve CD4+ T cells from young (Y) and older (O) individuals were activated for 5 days. Indicated inhibitor or vehicle control was added for the last 2 days of culture (A and B and E and F). Alternatively, cells were transfected with control or VPS39 siRNA and then activated with anti-CD3/anti-CD28 beads for 5 days (C and D). (A, C, E, and F) Lysosomal cathepsins expressions were determined by qRT-PCR (left); results are normalized to control samples using 18S rRNA as internal control; means ┬▒ SD of four experiments. Lysosomal activities were determined by flow cytometryтАУbased analysis of cells treated with DQ-BSA (5 ╬╝g/ml) for 6 hours. Results are shown as representative histograms (middle) and cumulative data from four experiments (right). The gray histogram represents DQ-BSA free samples. (B and D) Cells were treated with DQ-BSA (green) and stained with anti-pS6RP (red). Confocal images representative of two independent experiments show an inverse relationship between mTORC1 and lysosomal activity. Scale bars, 20 ╬╝m. Comparison by two-tailed paired t test (A, C, E, and F). *P < 0.05, **P < 0.01, and ***P < 0.001.
Sustained activation of late endosomal mTORC1 prevents PD-1 from lysosomal degradation
We previously observed that in the course of T cell proliferation after activation, FOXO1 promoted the generation of lysosomes through induction of TFEB transcription (27). Moreover, Foxo1-deficient mouse CD4+ T cell showed up to fourfold increase of PD-1 protein levels after antigen priming in vivo (39), raising the possibility that lysosomal activity regulates PD-1 protein expression. Lysosome inhibition by TFEB silencing in T cells from young adults increased cell surface protein expression of PD-1 while transcript levels remained unchanged (Fig. 4A). In contrast, stimulation of lysosomal activities after late endosomal mTORC1 inhibition in T cells from older adults by Torin 1 treatment, SLC7A5 inhibition, or VPS39 silencing reduced cell surface protein expression without effects on transcripts of PD-1 (Fig. 4, B to D). This effect was not due to the changes of intracellular versus cell surface trafficking routes of PD-1 since VPS39 silencing equally reduced cell surface and intracellular PD-1 protein expression (Fig. 4, E to G). Assessment of PD-1 half-life in vitro in day 5тАУstimulated older na├пve CD4+ T cells confirmed that VPS39-silenced cells underwent enhanced PD-1 protein degradation than control-silenced cells (Fig. 4H). These data suggest that mTORC1 at late endosomes stabilizes PD-1 protein by preventing lysosomal degradation of PD-1. In addition to PD-1, several molecules of functional relevance are targets of lysosomal degradation. cytotoxic T lymphocyteтАУassociated protein 4 (CTLA-4), another inhibitory receptor, was also reduced after enhancing lysosomal activities by late endosomal mTORC1 inhibition (fig. S5C).
(A to D) Na├пve CD4+ T cells from young and older individuals were activated with anti-CD3/anti-CD28 beads for 5 days with the last 2 days in the presence of vehicle or indicated inhibitor (B and C). Alternatively, cells were transfected with control or silencing RNA and activated for 5 days (A and D). Representative histograms showing cell surface protein expression of PD-1 (left) and cumulative data of cell surface protein expression (middle) and gene expression (right) of PD-1. (E to G) Cell surface expression (E), intracellular expression (F), and cell surface/intracellular PD-1 expression ratio (G) after control or VPS39 silencing in day 3тАУstimulated na├пve CD4+ T cells from older individuals. (H) Control or VPS39-silenced, day 5тАУstimulated na├пve CD4+ T cells from older individuals were treated with CHX (5 ╬╝g/ml) to inhibit de novo PD-1 synthesis. Total PD-1 protein normalized to ╬▓-actin expression is shown as relative to nontreatment. Means ┬▒ SEM of three experiments. (I) Longitudinal analysis of cell surface protein expression of PD-1 in na├пve CD4+ T cells from 10 young and 10 older individuals. Means ┬▒ SEM. (J) PD-1 gene expression comparison between day 5тАУstimulated young and older na├пve CD4+ T cells. The gray histogram represents isotype control. Comparison by two-tailed paired (A to G) or two-tailed unpaired t test (I and J). *P < 0.05, **P < 0.01, and ***P < 0.001.
To determine whether PD-1 protein regulation changes with age, we monitored PD-1 expression at days 0, 3, and 5 after stimulation by flow cytometry. Consistent with the changes of the kinetics of mTORC1 activity with age (8), PD-1 protein expression peaked at day 3 after activation in both young and older activated na├пve CD4+ T cells to then subside in activated T cells from young but not older adults. There were no age-related differences of protein expression of PD-1 at days 0 or 3, but activated T cells from older adults had more PD-1 protein at day 5 while no difference in PDCD1 transcription (Fig. 4, I and J). No age-related difference was seen for CTLA-4 expression, which occurred early in a T cell response and before the observed age-related differences in mTORC1 activity (fig. S6).