Abstract
NK cells are the first line of defense against infected and transformed cells. Defective NK cell activity was shown to increase susceptibility for viral infections and reduce tumor immune-surveillance. With age, the incidence of infectious diseases and malignancy rises dramatically, suggesting that impaired NK cell function might contribute to disease in these individuals. We found an increased frequency of NK cells with high expression of the inhibitory killer cell lectin-like receptor G1 (KLRG1) in individuals >70 y. The role of KLRG1 in ageing is not known, and the mechanism of KLRG1-induced inhibition of NK cell function is not fully understood. We report that NK cells with high KLRG1 expression spontaneously activate the metabolic sensor AMP-activated protein kinase (AMPK) and that activation of AMPK negatively regulates NK cell function. Pre-existing AMPK activity is further amplified by ligation of KLRG1 in these cells, which leads to internalization of the receptor and allows interaction with AMPK. We show that KLRG1 activates AMPK by preventing its inhibitory dephosphorylation by protein phosphatase-2C rather than inducing de novo kinase activation. Finally, inhibition of KLRG1 or AMPK prevented KLRG1-induced activation of AMPK and reductions in NK cell cytotoxicity, cytokine secretion, proliferation, and telomerase expression. This novel signaling pathway links metabolic sensing, effector function, and cell differentiation with inhibitory receptor signaling that may be exploited to enhance NK cell activity during ageing.
Introduction
The increase in human life expectancy is associated with a greater incidence and severity of many infections and malignancy in older subjects (1–4). The identification of mechanisms that are responsible for the immune decline is therefore essential for the rationalization of ways to improve health during later life. The expansion of T lymphocytes after immune activation takes a finite time, resulting in a lag phase before sufficient numbers of T cells are generated (5). During this period, the host is vulnerable to infections that spread rapidly and/or cause severe pathology, and this is prevented by NK cells (6). NK cells are a first line of defense against viral infections through their cytotoxic activity and ability to secrete cytokines without prior activation (7, 8). Individuals with rare genetic NK cell defects are susceptible to lethal herpes virus infections early postinfection but recover when optimal T cell responses are mobilized 1–2 wk later (9). Therefore, the efficient function of both T and NK cells is required to combat infection at different phases of the immune response. Although NK cell numbers generally increase during ageing, with a shift from an undifferentiated CD56bright to a differentiated CD56dim phenotype (10–13), these cells have reduced cytotoxicity, as well as a decreased capacity to secrete cytokines, such as IFN-γ, MIP-1α, and IL-8 (14). This reduction in NK cell function may lead to decreased immunity, especially during the early stages of infection in older subjects (6), as well as to an increased susceptibility and greater severity of viral infections in this population.
NK cell function is determined by a balance of activating and inhibitory signals delivered by activating receptors, which recognize stress-induced ligands, and inhibitory receptors, which engage MHC class I or MHC class–like molecules on healthy cells, respectively (15, 16). Killer cell lectin-like receptor G1 (KLRG1) is a C-type lectin-like inhibitory receptor with an immune receptor tyrosine-based inhibitory motif in its cytoplasmic domain (17). It binds to the ubiquitously expressed cell adhesion molecules E-cadherin, N-cadherin, and R-cadherin (18), and binding of E-cadherin to KLRG1 prevents lysis of E-cadherin–expressing target cells (18). In humans, KLRG1 is expressed by 50–80% of NK cells and 20–40% of T cells (19). Expression of the receptor is found on mature (CD56dim) NK cells (19, 20) and terminally differentiated T cells (18, 21, 22). Although KLRG1 expression on T cells was shown to dramatically increase with age (21–25), data on KLRG1 expression on NK cells in elderly individuals is scarce and controversial (26, 27). In a study by Hayhoe et al. (27), KLRG1 expression was found to be decreased, instead of increased, in old subjects. However, unlike in the present investigation, in which all of the samples were freshly isolated, the study by Hayhoe et al. used a mixture of fresh and frozen samples. Recent data from our group suggest that KLRG1 expression is reduced considerably during cryopreservation, which would explain the discrepancy between the data (S.M. Henson, unpublished observations). High KLRG1 expression correlates with low proliferative capacity (28, 29), impaired IFN-γ secretion (29, 30), and increased apoptosis (29) in NK cells. Moreover, in patients chronically infected with hepatitis C, blockade of KLRG1 signaling restored defective protein kinase B (Akt) phosphorylation and IFN-γ secretion (29), suggesting that KLRG1 actively contributes to functional defects observed in KLRG1+ cells.
AMP-responsive protein kinase (AMPK) is a protein sensor that integrates intracellular cues, such as low ATP levels and DNA damage signals, to regulate cell function (31, 32). We previously identified a novel AMPK-dependent pathway in highly differentiated CD4+ T cells that is activated by senescence and low nutrient sensing signals to inhibit proliferation and telomerase activity (32). Once active, AMPK triggers p38 MAPK activation, and the inhibition of either molecule restores proliferation and telomerase activity (32). Because terminally differentiated NK cells with impaired immune function accumulate during ageing (14), we hypothesized that the decline in NK cell function during ageing may also be regulated by an analogous AMPK-dependent mechanism. We found that, independently of its canonical inhibitory target Akt (22, 29), KLRG1 stimulated AMPK activity in NK cells with high KLRG1 expression (KLRG1bright), and signaling through this pathway inhibited NK cell cytotoxicity, IFN-γ production, proliferation, and telomerase activity. Furthermore, KLRG1 was internalized upon ligation, bound to AMPK directly, and prevented its inhibitory dephosphorylation by protein phosphatase-2C (PP2C) phosphatases. This is a hitherto unrecognized mechanism of cell surface inhibitory receptor function in lymphocytes in which AMPK activation is amplified through protection from dephosphorylation, rather than de novo kinase activation. The central implication of these observations is that inhibition of KLRG1/AMPK signaling may restore NK cell function that is relevant for immune enhancement during ageing and in patients with malignancy.
Materials and Methods
Study population and blood sample collection
After written informed consent was obtained, heparinized peripheral blood samples were taken from healthy volunteers without previous record of an autoimmune or malignant disease. Where donors are stratified by age, young is defined as ≤35 y old (age range: 20–34 y, median: 27 y, mean: 25.8 y) and old is defined as ≥70 y of age (age range: 70–86 y, median: 75 y, mean: 76.9 y).
Cell isolation
PBMCs were isolated from heparinized peripheral blood using Ficoll-Paque (Amersham Biosciences). NK cells were isolated from PBMCs with the NK Cell Isolation Kit (Miltenyi Biotec), according to the manufacturer’s instructions. To obtain NK cell subsets with differential KLRG1 expression, NK cells were labeled with a biotinylated anti-KLRG1 Ab (BioLegend), washed, and labeled with Anti-Biotin MicroBeads (Miltenyi Biotec), according to the manufacturer’s instructions. KLRG1bright NK cells were obtained by passing KLRG1-labeled NK cells through a magnetic column (Miltenyi Biotec) where KLRG1bright NK cells were retained (positive fraction). Flow-through was run over two additional magnetic columns sequentially to obtain KLRG1dim NK cells (positive fraction). All remaining cells were considered KLRG1neg (negative fraction). Cell surface KLRG1 expression after cell isolation was determined with Streptavidin-Cy5 (BioLegend) or a directly labeled anti-KLRG1 Ab (BioLegend) (Supplemental Fig. 1B).
Cell culture
NK cells were cultured in complete medium (RPMI 1640 supplemented with 10% heat-inactivated FCS, 100 U/ml Penicillin, 100 mg/ml Streptomycin, and 2 mM l-glutamine; all from Invitrogen) unless otherwise stated. Glucose-free RPMI 1640 (Life Technologies) was used for glucose-deprivation experiments (6 h or overnight). MCF-10A cells were cultured in DMEM/F12 medium (Invitrogen) containing 5% horse serum (Life Technologies), 20 ng/ml epidermal growth factor (PeproTech), 0.5 mg/ml Hydrocortisone, 100 ng/ml Cholera Toxin, 10 μg/ml Insulin (all from Sigma-Aldrich), 100 U/ml Penicillin, and 100 mg/ml Streptomycin (both from Invitrogen).
Flow cytometry
The following Abs (all from BioLegend, unless otherwise indicated) were used: anti-KLRG1–biotin (2F1), anti-KLRG1–PE (2F1), anti-CD3 (UCHT1; BD Biosciences), anti-CD56 (HCD56), anti-CD16 (3G8; BD Biosciences), and anti-CD7 (M-T701). Biotin-conjugated Abs were detected using Cy5- or Cy3-conjugated Streptavidin (BioLegend). Annexin V staining was performed using Annexin V Binding Buffer and Annexin V (both from BioLegend). Ki67 staining was performed (B56; BD Biosciences) with the Foxp3 Staining Set (Miltenyi Biotec), according to the manufacturer’s instructions. When using total PBMCs, NK cells were identified as CD3−CD7+ lymphocytes (33) (Supplemental Fig. 1A). All samples were acquired on an LSR II flow cytometer (BD Biosciences). Data were analyzed using FlowJo v10 software (TreeStar, Ashland, OR).
Phospho-flow cytometry
After cell surface staining on ice, cells were fixed with Cytofix Buffer (BD Biosciences) for 10 min at 37°C and permeabilized with ice-cold Perm Buffer III (BD Biosciences), followed by staining with the following Abs for 30 min at room temperature: anti–p-AMPKα (Thr172) rabbit mAb (40H9), anti–p-ATM (Ser1981) rabbit mAb (D6H9), anti–γH2Ax-PE (Ser139) mouse mAb (20E3) (all from Cell Signaling Technology), and anti–p-Akt (Ser473) mouse mAb (M89-61; BD Biosciences). Primary Abs were detected with goat anti-rabbit, goat anti-mouse, or donkey anti-rabbit IgG (all from Life Technologies) for 30 min at room temperature. All samples were acquired immediately after the staining on an LSR II flow cytometer (BD Biosciences). Data were analyzed using FlowJo v10 software (TreeStar).
Calcein-release cytotoxicity assay
The cytotoxic capacity of NK cells was assessed using the E-cadherin–expressing breast cancer epithelial cell line MCF-10A as target. Briefly, 20,000 MCF-10A cells were plated/well of a 96-well flat-bottom plate 24 h prior to the cytotoxicity assay. On the day of the assay, MCF-10A cells were labeled with Calcein-AM (Sigma-Aldrich) at 10 μM for 1 h before coculture with NK cells in complete medium containing 500 IU/ml recombinant human (rh)IL-2 (Miltenyi Biotec) and as described (34). NK cells transfected with small interfering RNA (siRNA) specific for KLRG1 or scrambled control siRNA for 36 h were pretreated with the AMPK agonist A769662 (Tocris Bioscience) at 150 μM or equivalent DMSO as negative control for 2 h before being added to the target cells. Effector and target cells were combined at a ratio of 40:1 in triplicate and cultured in complete medium containing 500 IU/ml rhIL-2 (Miltenyi Biotec) for 4 h. After 4 h of coculture, fluorescence was measured in 75 μl of cell culture supernatant using a SpectraMax Gemini spectrofluorometer. Specific lysis was calculated as percentage killing: (test release − spontaneous release)/(max release − spontaneous release) × 100.
Granzyme B and IFN-γ analysis
For quantification of granzyme B and IFN-γ in cell culture supernatants, a Cytometric Bead Array Assay (Human Granzyme B and Human IFN-γ Flex Set; BD Biosciences) was used. Measurements were performed according to the manufacturer’s instructions. The lower limit of detection was 4 pg/ml for granzyme B and 0.8 pg/ml for IFN-γ.
Introduction of siRNA into primary human NK cells
siRNA was introduced into freshly isolated KLRG1bright NK cells by electroporation using the Amaxa Human NK Cell Nucleofector Kit and Nucleofector technology (Lonza), according to the manufacturer’s instructions. Briefly, 3 × 106 freshly isolated KLRG1bright NK cells were resolved in Nucleofector solution provided by the manufacturer and 200 nmol PP2C (PP2Cα siRNA; sc-42937) or 300 nmol KLRG1 siRNA (sc-42937; both from Santa Cruz Biotechnology). Nucleofection was performed with the Nucleofector I Device Program U-01 (Lonza). Cells were transferred into 12-well cell culture plates containing pre-equilibrated complete medium and incubated for 36 h. NK cells transfected with KLRG1 siRNA were cultured in complete medium containing 500 IU/ml rhIL-2 (Miltenyi Biotec). Knockdown efficiency of KLRG1 siRNA was monitored by measuring cell surface expression of KLRG1 by flow cytometry (Supplemental Fig. 3A). Knockdown of PP2Cα was confirmed by immunoblot analysis (rabbit anti-PP2Cα mAb, clone D18C10, XP; Cell Signaling Technology) (Supplemental Fig. 3B). A scrambled control siRNA (sc-37007; Santa Cruz Biotechnology) was used in all experiments.
Lentiviral transduction of primary human NK cells
Freshly isolated KLRG1bright NK cells were preactivated with K562 target cells (E:T ratio of 10:1) and 200 IU rhIL-2 (Miltenyi Biotec) for 48 h. On day 2, cells were transduced with pHIV1-Siren lentiviral particles, as previously described (32). On day 6 of culture, cells were transferred to a 96-well plate coated with an anti-KLRG1 mAb or an IgG control Ab (both at 10 μg/ml; both from BioLegend), and K562 target cells (E:T ratio of 10:1) and fresh IL-2 (200 IU/ml) were added. On day 9 of culture, apoptosis was assessed by Annexin V staining, and proliferation was measured by Ki67 staining. Human telomerase reverse transcriptase (hTERT) expression was measured by intranuclear staining with the anti-TERT mAb (ab68781; Abcam). On day 9, NK cells were restimulated with K562 target cells at an E:T ratio of 10:1 and fresh IL-2 (200 IU/ml) and cultured for a total of 12 d (for experimental design see Supplemental Fig. 4A).
Immunoprecipitation
For coimmunoprecipitation of AMPK, PP2C, and KLRG1, cell lysate from 10 × 106 isolated KLRG1bright NK cells was prepared as previously described (32). Cell lysate was split into two and incubated with a polyclonal anti-AMPKα Ab or an isotype-control Ab (both from Cell Signaling Technology). Immunoprecipitated proteins were detected by immunoblot analysis with an anti-KLRG1 (ab170959; Abcam), anti-PP2C and anti-AMPK Ab (both from Cell Signaling Technology), and confirmation-specific mouse anti-rabbit IgG Ab (L27A9; Cell Signaling Technology), a mouse Ab to rabbit IgG L chain (L57A3; Cell Signaling Technology), or a secondary anti-mouse IgG Ab (7076; Cell Signaling), and developed with the ECL Prime Western detection kit (GE Healthcare).
In vitro phosphatase assay
AMPK immunoprecipitates were obtained as described above. After extensive washing, PP2C-like (PP2ase) activity was measured using the ProFluor Ser/Thr PPase Assay, according to the manufacturer’s instructions (Promega), and normalized against total levels of immunoprecipitated AMPK. Immunoprecipitated AMPK was detected by ELISA-based assays (ab181422; Abcam), according to the manufacturer’s instructions.
ImageStream flow cytometry
Purified KLRG1dim and KLRG1bright NK cells were incubated with AMPK agonist A-769662 (150 μM) or DMSO as a negative control for 12 h, followed by KLRG1 ligation or an isotype-control Ab (both from BioLegend) for 2 h at 37°C. Subsequently, cells were fixed with 2% paraformaldehyde at 37°C for 10 min and permeabilized with ice-cold Perm Buffer III (BD Biosciences) for 30 min on ice. KLRG1 was stained with a biotinylated anti-KLRG1 mAb, followed by Streptavidin-Cy5 staining (both from BioLegend). p-AMPKα was stained with an anti–p-AMPKα (Thr172) rabbit mAb and goat anti-rabbit IgG (both from Cell Signaling Technology). Samples were run on an Amnis ImageStream cytometer using INSPIRE software (magnification ×60). Data were compensated and analyzed using IDEAS v.6.1 software (Amnis). Colocalization of KLRG1 and p-AMPK was determined on a single-cell basis using Bright Detail Similarity (BDS) score analysis. Colocalization was considered as BDS ≥ 2.0.
Statistics
GraphPad Prism software was used to perform all statistical analyses. For parametric data, the Student t test or repeated-measures ANOVA test with Greenhouse–Geisser correction was used. For nonparametric data, the Wilcoxon matched-pairs signed rank test or Friedman test was used. The p values <0.05 were considered significant.
Ethical approval
The present study was approved by the Ethical Committee of the Royal Free and University College London Medical School. Written informed consent was obtained from all study participants.
Results
KLRG1bright NK cells accumulate with age
On the basis of their relative expression of CD56 and CD16, we identified three subsets of human NK cells in peripheral blood: CD56dim (CD56+CD16++, 80.5 ± 7.2%), CD56bright (CD56++CD16−, 4.6 ± 2.8%), and CD56brightCD16+ (CD56++CD16+, 2.39 ± 1.2%) (Fig. 1A, Supplemental Fig. 1A). When comparing relative NK cell numbers between young (≤35 y) and old (≥70 y) donors, we found, consistent with reports from other groups (13, 35, 36), that the percentage of CD56dim NK cells was significantly increased in older individuals, whereas the percentage of the more immature CD56bright and CD56brightCD16+ NK cell subsets was reduced compared with young donors (Fig. 1B). When comparing expression of KLRG1 on NK cell subsets in young and old donors, we found that, irrespective of age, KLRG1 expression was predominant within the more differentiated CD56dim NK cell subset (Fig. 1C). Moreover, old donors had an increase in the frequency of KLRG1-expressing NK cells compared with young donors, and this was true for the CD56brightCD16+ and CD56dim NK cell subsets (Fig. 1C). In addition to an increase in KLRG1pos NK cells, we found a higher expression level of KLRG1 in NK cells from old donors compared with young donors (Fig. 1D). Because the inhibitory capacity of KLRG1 is known to be directly proportional to the cell surface expression level of the receptor (37), we stratified CD56dim NK cells, the main NK cell population expressing KLRG1, into cells with no (KLRG1neg), intermediate (KLRG1dim), or high (KLRG1bright) KLRG1 expression levels (Fig. 1D). Using this approach, we found a significant increase in the percentage of KLRG1bright NK cells in old donors compared with young donors (Fig. 1E). This was most pronounced within the CD56dim NK cell compartment, whereas KLRG1 expression was low in CD56bright NK cells of both age groups (Fig. 1C, 1E).
KLRG1bright NK cells accumulate with age. (A) NK cell subsets in peripheral blood stratified by CD56 and CD16 expression into CD56bright (CD56++CD16−), CD56brightCD16+, and CD56dim (CD56+CD16++) NK cells from one representative young donor (≤35 y). (B) Cumulative data from 15 young donors (≤35 y) and 15 old donors (≥70 y) analyzed as in (A). (C) Percentages of KLRG1+ NK cells within each NK cell subset, defined as in (A), as assessed in 15 young and 15 old donors. (D) CD56dim NK cells were divided into cells expressing no (KLRG1neg), intermediate (KLRG1dim), and high levels (KLRG1bright) of the receptor, indicated by −, +, and ++, respectively. One representative young and one representative old donor are shown. Numbers indicate percentages of KLRG1neg, KLRG1pos, and KLRG1bright NK cells in total CD56dim NK cells. (E) Percentage of KLRG1bright NK cells in each NK cell subset, as defined in (A), in 15 young and 15 old donors. All experiments were performed on total PBMCs, applying the gating strategy illustrated in Supplemental Fig. 1A. All bars represent mean values ± SEM. The p values were calculated using the unpaired Student t test. **p ≤ 0.01, ***p ≤ 0.001. ns, p > 0.05.
KLRG1bright NK cells accumulate with age. (A) NK cell subsets in peripheral blood stratified by CD56 and CD16 expression into CD56bright (CD56++CD16−), CD56brightCD16+, and CD56dim (CD56+CD16++) NK cells from one representative young donor (≤35 y). (B) Cumulative data from 15 young donors (≤35 y) and 15 old donors (≥70 y) analyzed as in (A). (C) Percentages of KLRG1+ NK cells within each NK cell subset, defined as in (A), as assessed in 15 young and 15 old donors. (D) CD56dim NK cells were divided into cells expressing no (KLRG1neg), intermediate (KLRG1dim), and high levels (KLRG1bright) of the receptor, indicated by −, +, and ++, respectively. One representative young and one representative old donor are shown. Numbers indicate percentages of KLRG1neg, KLRG1pos, and KLRG1bright NK cells in total CD56dim NK cells. (E) Percentage of KLRG1bright NK cells in each NK cell subset, as defined in (A), in 15 young and 15 old donors. All experiments were performed on total PBMCs, applying the gating strategy illustrated in Supplemental Fig. 1A. All bars represent mean values ± SEM. The p values were calculated using the unpaired Student t test. **p ≤ 0.01, ***p ≤ 0.001. ns, p > 0.05.
KLRG1 activates AMPK signaling in KLRG1bright NK cells
We showed previously that AMPK is spontaneously active (Thr172 phosphorylation in the AMPK-activation loop) in highly differentiated/senescent T cells (32), and we questioned whether this also occurred in CD56dim NK cells that accumulate with age. We gated on the three NK cell populations, as defined in Fig. 1A, and found that AMPK was only active in the CD56dim NK cell subset (Fig. 2A, 2B). Because these cells also express high levels of KLRG1 (Fig. 1C–E), we investigated whether both molecules were detectable within the same cell (Fig. 2C). When we isolated NK cells on the basis of negative, dim, or bright KLRG1 expression using magnetic beads (Supplemental Fig. 1B), we found that only KLRG1bright NK cells exhibited spontaneous AMPK activation (Fig. 2C, 2D). Functional analysis of KLRG1bright NK cells revealed low proliferative activity (Supplemental Fig. 2A), telomere shortening (Supplemental Fig. 2B), and low expression of the catalytic component of telomerase (hTERT; Supplemental Fig. 2C), as well as endogenous phosphorylation of the DNA damage response protein H2AX (γH2Ax; Supplemental Fig. 2D) and its upstream activator, ataxia telangiectasia mutated protein (p-ATM; Supplemental Fig. 2E), compared with KLRG1dim and KLRG1neg NK cells. These data indicate that KLRG1bright cells express multiple characteristics of cellular senescence (38, 39) and suggest that activation of AMPK in KLRG1bright NK cells may be derived from cell senescence signals (32).
KLRG1 activates AMPK signaling in KLRG1bright NK cells. (A) CD56dim NK cells spontaneously activate AMPK (Thr172). Total NK cells isolated directly ex vivo from one representative old donor, stratified to CD56 and CD16 expression (left panel). Only NK cells that spontaneously activate AMPK (AMPK phosphorylation at Thr172 assessed directly ex vivo) within total NK cells isolated directly ex vivo from the same donor stratified to CD56 and CD16 (right panel). The gate indicates CD56dim NK cells. (B) Cumulative data from 10 donors showing spontaneous AMPK phosphorylation (Thr172) in NK cell subsets within total PBMCs, as described in Supplemental Fig. 1A. Data are shown as fold change in mean fluorescence intensity (MFI) of p-AMPK normalized to the CD56bright population. (C) Phosphorylation of AMPK (Thr172), as analyzed directly ex vivo in purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells from the same representative donor. Numbers indicate MFI of p-AMPK (Thr172). (D) Cumulative data from 10 donors showing AMPK phosphorylation in KLRG1bright, KLRG1dim, and KLRG1neg NK cells, as assessed directly ex vivo in total PBMCs. Data are shown as fold change in MFI of p-AMPK (Thr172) normalized to KLRG1neg NK cells. KLRG1 ligation in freshly purified KLRG1neg, KLRG1dim, and KLRG1bright NK cell subsets enhances AMPK activity in KLRG1bright NK cells only (E), whereas Akt inhibition is only seen in KLRG1dim cells (F). Pooled data from five independent experiments, all performed on freshly isolated KLRG1 subsets. (G) Akt expression in freshly purified KLRG1neg, KLRG1dim, and KLRG1bright NK cell subsets, as assessed by intracellular staining. Representative graph from one of three donors tested. (H) Knockdown of KLRG1 by siRNA reduces AMPK phosphorylation in KLRG1bright, but not in KLRG1dim, NK cells compared with control siRNA. Data from one representative donor, as assessed on freshly purified KLRG1bright and KLRG1dim NK cells. (I) Cumulative data from four independent experiments analyzed as in (H). Bars represent mean values ± SEM (B, D, E, and F) and median values ± interquartile range (I). The p values were calculated using the Student t test for parametric data and the Friedman test for nonparametric data. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. ns, p > 0.05.
KLRG1 activates AMPK signaling in KLRG1bright NK cells. (A) CD56dim NK cells spontaneously activate AMPK (Thr172). Total NK cells isolated directly ex vivo from one representative old donor, stratified to CD56 and CD16 expression (left panel). Only NK cells that spontaneously activate AMPK (AMPK phosphorylation at Thr172 assessed directly ex vivo) within total NK cells isolated directly ex vivo from the same donor stratified to CD56 and CD16 (right panel). The gate indicates CD56dim NK cells. (B) Cumulative data from 10 donors showing spontaneous AMPK phosphorylation (Thr172) in NK cell subsets within total PBMCs, as described in Supplemental Fig. 1A. Data are shown as fold change in mean fluorescence intensity (MFI) of p-AMPK normalized to the CD56bright population. (C) Phosphorylation of AMPK (Thr172), as analyzed directly ex vivo in purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells from the same representative donor. Numbers indicate MFI of p-AMPK (Thr172). (D) Cumulative data from 10 donors showing AMPK phosphorylation in KLRG1bright, KLRG1dim, and KLRG1neg NK cells, as assessed directly ex vivo in total PBMCs. Data are shown as fold change in MFI of p-AMPK (Thr172) normalized to KLRG1neg NK cells. KLRG1 ligation in freshly purified KLRG1neg, KLRG1dim, and KLRG1bright NK cell subsets enhances AMPK activity in KLRG1bright NK cells only (E), whereas Akt inhibition is only seen in KLRG1dim cells (F). Pooled data from five independent experiments, all performed on freshly isolated KLRG1 subsets. (G) Akt expression in freshly purified KLRG1neg, KLRG1dim, and KLRG1bright NK cell subsets, as assessed by intracellular staining. Representative graph from one of three donors tested. (H) Knockdown of KLRG1 by siRNA reduces AMPK phosphorylation in KLRG1bright, but not in KLRG1dim, NK cells compared with control siRNA. Data from one representative donor, as assessed on freshly purified KLRG1bright and KLRG1dim NK cells. (I) Cumulative data from four independent experiments analyzed as in (H). Bars represent mean values ± SEM (B, D, E, and F) and median values ± interquartile range (I). The p values were calculated using the Student t test for parametric data and the Friedman test for nonparametric data. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. ns, p > 0.05.
We next examined the effects of KLRG1 ligation on AMPK activity in purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells. We used phosphorylation of Akt (Ser473), an established KLRG1-inhibitory target (22, 29), as a readout control. The addition of an agonistic KLRG1 Ab to freshly isolated KLRG1neg, KLRG1dim, or KLRG1bright NK cells robustly enhanced AMPK activity in KLRG1bright NK cells but had no effect on the KLRG1dim or KLRG1neg population (Fig. 2E). In contrast, Akt phosphorylation was not altered in response to KLRG1 ligation in KLRG1bright or KLRG1neg NK cells but was inhibited in KLRG1dim cells (Fig. 2F). No change in AMPK or Akt activity could be detected when an irrelevant isotype-control Ab was used (Fig. 2E, 2F). Correspondingly, there was relatively low Akt expression in freshly purified KLRG1bright NK cells that exhibit endogenous AMPK activity (Fig. 2G versus 2C) compared with KLRG1dim and KLRG1neg NK cells.
To corroborate these findings, we silenced KLRG1 by siRNA in ex vivo–isolated KLRG1bright and KLRG1dim NK cells (Supplemental Fig. 3A). Knockdown of KLRG1 reduced AMPK phosphorylation in KLRG1bright NK cells but had no significant effect on KLRG1dim cells that lack endogenous AMPK phosphorylation (Fig. 2H, 2I). Together, these findings show that KLRG1 activates AMPK signaling in highly differentiated primary human NK cells. Furthermore, Akt inhibition and AMPK activation constitute two independent pathways of KLRG1 function that may operate at different stages of NK cell differentiation.
KLRG1 activates AMPK by preventing its dephosphorylation
Because KLRG1 ligation only enhanced AMPK phosphorylation in NK cells with pre-existing (endogenous) AMPK activity, we reasoned that KLRG1 engagement may activate AMPK through inhibition of dephosphorylation. In ex vivo–isolated KLRG1 subsets, ligation of KLRG1 induced AMPK activation only in KLRG1bright NK cells (Fig. 3A). When we cultured purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells in glucose-free medium (40), AMPK phosphorylation was detected in all three cell subsets (Fig. 3A). In glucose-starved NK cells, KLRG1 engagement with the activating KLRG1 Ab enhanced AMPK activity not only in KLRG1bright cells but also in KLRG1dim cells by 2–3-fold (Fig. 3A). There were no KLRG1-related changes in AMPK activation in KLRG1neg NK cells (Fig. 3A). This indicates that KLRG1 ligation enhances pre-existing AMPK activity. Therefore, we reasoned that KLRG1 might regulate AMPK function by preventing its inhibitory dephosphorylation by upstream phosphatases rather than directly inducing de novo AMPK activation.
KLRG1 activates AMPK by preventing its dephosphorylation. (A) Pre-existing AMPK activity is required for KLRG1 regulation of AMPK. Purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells were left untreated or cultured in glucose-free medium for 6 h, followed by KLRG1 ligation (n = 5 independent experiments). An isotype Ab was used as negative control. (B) Phosphorylation of AMPK in purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells transfected with siRNA to PP2Cα (siPP2C) or control siRNA (siCtrl) for 36 h, followed by a 2-h stimulation with the activating KLRG1 Ab or medium alone (n = 3 independent experiments). (C) Activity of PP2C-like phosphatases in AMPK immune-precipitates from KLRG1bright NK cells stimulated with an activating KLRG1 mAb or an isotype-control Ab (2 h, 37°C). Phosphatase activity was normalized to immunoprecipitated AMPK by ELISA (n = 3). Bars represent median values ± interquartile range. The p values were calculated using a Friedman test for nonparametric data. *p ≤ 0.05, **p ≤ 0.01. ns, p > 0.05.
KLRG1 activates AMPK by preventing its dephosphorylation. (A) Pre-existing AMPK activity is required for KLRG1 regulation of AMPK. Purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells were left untreated or cultured in glucose-free medium for 6 h, followed by KLRG1 ligation (n = 5 independent experiments). An isotype Ab was used as negative control. (B) Phosphorylation of AMPK in purified KLRG1neg, KLRG1dim, and KLRG1bright NK cells transfected with siRNA to PP2Cα (siPP2C) or control siRNA (siCtrl) for 36 h, followed by a 2-h stimulation with the activating KLRG1 Ab or medium alone (n = 3 independent experiments). (C) Activity of PP2C-like phosphatases in AMPK immune-precipitates from KLRG1bright NK cells stimulated with an activating KLRG1 mAb or an isotype-control Ab (2 h, 37°C). Phosphatase activity was normalized to immunoprecipitated AMPK by ELISA (n = 3). Bars represent median values ± interquartile range. The p values were calculated using a Friedman test for nonparametric data. *p ≤ 0.05, **p ≤ 0.01. ns, p > 0.05.
Dephosphorylation of AMPK by upstream phosphatases, primarily PP2C, represents an important mechanism of AMPK regulation (41, 42). We tested whether KLRG1 enhanced AMPK activity through inhibition of PP2C. Immunoblot analysis revealed comparable PP2C expression levels in all three KLRG1 subsets (data not shown). We then silenced PP2C by siRNA (Supplemental Fig. 3B) and found that KLRG1 ligation failed to enhance AMPK activity in KLRG1bright NK cells in the absence of PP2C (Fig. 3B). Of note, PP2C knockdown did not alter KLRG1 expression or cell viability in these cells (data not shown), indicating that the unresponsiveness of PP2C-silenced NK cells to KLRG1 ligation was not due to a reduction in KLRG1 cell surface expression. Thus, KLRG1 activates AMPK through a PP2C-dependent mechanism.
To assess whether KLRG1 signaling modulates activity of PP2C-like AMPK-associated phosphatases, we ligated KLRG1 on KLRG1bright NK cells, immunoprecipitated AMPK, and performed ELISA-based in vitro phosphatase assays. We found that KLRG1 ligation significantly reduced phosphatase activity in AMPK immunoprecipitates compared with IgG control ligation (Fig. 3C). These data show that KLRG1 signaling impedes dephosphorylation of AMPK, a process mediated by the phosphatase PP2C.
KLRG1 interacts with AMPK and undergoes internalization
We immunoprecipitated AMPK from freshly isolated KLRG1bright NK cells and found that KLRG1, AMPK, and PP2C interacted endogenously (Fig. 4A). Because KLRG1 signaling inhibits phosphatase activity in the AMPK complex (Fig. 3C), we reasoned that KLRG1 ligation may disrupt AMPK–PP2C interaction. When we immunoprecipitated AMPK from KLRG1dim NK cells treated with the AMPK agonist A-769662 or DMSO vehicle control, we found that KLRG1 ligation dissociated PP2C from AMPK, a process that was most evident on an AMPK-activation background (Fig. 4B, lanes 3 and 4).
KLRG1 interacts with AMPK and undergoes internalization. (A) AMPK, PP2C, and KLRG1 interact endogenously. KLRG1bright NK cells were lysed, immunoprecipitated with AMPK or IgG control Ab, and subjected to immunoblotting. Representative of two separate experiments. (B) KLRG1 signaling disrupts AMPK-PP2C interaction. Isolated KLRG1dim NK cells were cultured with the AMPK agonist A-769662 (150 μM) or DMSO as vehicle control overnight, with or without KLRG1 ligation for 2 h on the following day. AMPK-bound PP2C was detected in AMPK immunoprecipitates by immunoblotting. Whole-cell lysates (WL) of PP2C protein served as loading control throughout. Representative of two separate experiments. (C) KLRG1 downregulation in purified KLRG1bright NK cells cultured overnight in complete medium and then stimulated with an activating KLRG1 Ab (2 h, 37°C; red line) or the AMPK agonist A-769662 (150 μM overnight; blue line) or cultured in glucose-free medium (overnight; green line) compared with KLRG1bright NK cells cultured overnight in complete medium only (black line). Mean fluorescence intensity values of cell surface KLRG1 are shown. (D) ImageStream analysis of p-AMPK and KLRG1 colocalization in purified KLRG1dim and KLRG1bright NK cells (original magnification ×60). Colocalization occurs endogenously in KLRG1bright NK cells that spontaneously activate AMPK [(1C) DSMO] but requires exogenous AMPK activity in the KLRG1dim population [(1C) AMPK act]. AMPK was activated by A-769662 (150 μM, overnight), with or without KLRG1 ligation. A DMSO vehicle solution was used as control. (E) Cumulative data from two independent experiments for KLRG1dim and KLRG1bright NK cells, indicating the percentages of NK cells positive for p-AMPK–KLRG1 colocalization (BDS > 2) and shown as fold change compared with untreated NK cells.
KLRG1 interacts with AMPK and undergoes internalization. (A) AMPK, PP2C, and KLRG1 interact endogenously. KLRG1bright NK cells were lysed, immunoprecipitated with AMPK or IgG control Ab, and subjected to immunoblotting. Representative of two separate experiments. (B) KLRG1 signaling disrupts AMPK-PP2C interaction. Isolated KLRG1dim NK cells were cultured with the AMPK agonist A-769662 (150 μM) or DMSO as vehicle control overnight, with or without KLRG1 ligation for 2 h on the following day. AMPK-bound PP2C was detected in AMPK immunoprecipitates by immunoblotting. Whole-cell lysates (WL) of PP2C protein served as loading control throughout. Representative of two separate experiments. (C) KLRG1 downregulation in purified KLRG1bright NK cells cultured overnight in complete medium and then stimulated with an activating KLRG1 Ab (2 h, 37°C; red line) or the AMPK agonist A-769662 (150 μM overnight; blue line) or cultured in glucose-free medium (overnight; green line) compared with KLRG1bright NK cells cultured overnight in complete medium only (black line). Mean fluorescence intensity values of cell surface KLRG1 are shown. (D) ImageStream analysis of p-AMPK and KLRG1 colocalization in purified KLRG1dim and KLRG1bright NK cells (original magnification ×60). Colocalization occurs endogenously in KLRG1bright NK cells that spontaneously activate AMPK [(1C) DSMO] but requires exogenous AMPK activity in the KLRG1dim population [(1C) AMPK act]. AMPK was activated by A-769662 (150 μM, overnight), with or without KLRG1 ligation. A DMSO vehicle solution was used as control. (E) Cumulative data from two independent experiments for KLRG1dim and KLRG1bright NK cells, indicating the percentages of NK cells positive for p-AMPK–KLRG1 colocalization (BDS > 2) and shown as fold change compared with untreated NK cells.
When we activated AMPK in purified KLRG1bright NK cells by KLRG1 ligation, glucose-deprivation, or selective AMPK agonist stimulation (43), we observed a consistent reduction in cell surface KLRG1 expression (Fig. 4C). We reasoned that KLRG1 might have been internalized to facilitate interaction with AMPK. To test for internalization of KLRG1, we compared KLRG1dim NK cells that lack endogenous AMPK activation and KLRG1bright NK cells that spontaneously activate AMPK. To this end, we cultured isolated KLRG1dim and KLRG1bright NK cells overnight in the presence of the AMPK agonist A-769662 or DMSO vehicle control, followed by ligation of KLRG1 for 2 h and ImageStream analysis.
On a DMSO background, we found that KLRG1bright, but not KLRG1dim, NK cells showed endogenous AMPK activation and colocalization of KLRG1 and p-AMPK (Fig. 4D top panel, Fig. 4E). When KLRG1 was ligated, we found enhanced p-AMPK/KLRG1 colocalization in KLRG1bright cells only (Fig. 4D, second panel, Fig. 4E). In contrast, AMPK was refractory to triggering of the receptor per se in KLRG1dim NK cells (Fig. 4D second panel, Fig. 4E). Because pre-existing AMPK activity is essential for KLRG1 regulation of AMPK (see above), we reasoned that enforcing AMPK activation in KLRG1dim cells would reconstitute p-AMPK/KLRG1 colocalization, even in this less-differentiated population. On an AMPK-activation background, we indeed observed KLRG1 and p-AMPK colocalization in KLRG1bright NK cells, as well as in KLRG1dim NK cells (Fig. 4D third panel, Fig. 4E). KLRG1 ligation now enhanced p-AMPK/KLRG1 colocalization further in both cell types (Fig. 4D fourth panel, Fig. 4E). Quantification of colocalization is shown in Fig. 4E for all four conditions, and quantification of KLRG1 and p-AMPK signals is shown in Supplemental Fig. 3C. We obtained comparable results when probing colocalization of KLRG1 and total AMPK by ImageStream (data not shown). Together, these data support a model whereby KLRG1 internalizes upon ligation, an event that requires pre-existing AMPK activation, and further enhances AMPK signaling.
KLRG1-induced inhibition of NK cell function is AMPK dependent
We next investigated the functional implications of inhibiting NK cells via the KLRG1/AMPK axis. We probed KLRG1 function by coculturing KLRG1bright NK cells with the breast cancer cell line MCF-10A, which expresses abundant levels of the KLRG1 ligand E-cadherin (44). Knockdown of KLRG1 by siRNA increased killing capacity (measured by direct tumor cell lysis), as well as release of granzyme B and IFN-γ secretion of KLRG1bright NK cells, toward the MCF-10A cell line (Fig. 5A). Importantly, this process was AMPK dependent, because all three functions examined were abolished by addition of the AMPK agonist A-769662 to the KLRG1-silenced NK cells (Fig. 5A). These data identify an inhibitory pathway of KLRG1 function in primary human NK cells that requires AMPK signaling.
KLRG1-induced inhibition of NK cell function is AMPK dependent. (A) Cytotoxic activity assessed by calcein-release lysis assay in purified KLRG1bright NK cells transfected with siRNA specific for KLRG1 (siKLRG1) or control siRNA (siCtrl) for 36 h and tested vis-à-vis the E-cadherin–expressing tumor cell line MCF-10A (4 h coculture). Throughout the cell culture, NK cells were kept in complete medium supplemented with 500 IU/ml rhIL-2. Release of granzyme B and IFN-γ under the same conditions are also shown. Reliance of KLRG1 on AMPK activity was probed by addition of the AMPK agonist A-769662 (150 μM) to the transfected cells 2 h prior to the cytotoxicity assay. A DMSO solution was used as vehicle control. Data are pooled from six donors (specific lysis) or five donors (granzyme B and IFN-γ). (B–G) KLRG1 ligation negatively regulates NK cell activity in AMPK-proficient cells only. Purified KLRG1bright NK cells were transduced with lentiviral vectors containing silencing shAMPK or shCtrl, as described. On days 6 and 9, NK cells were restimulated with feeder cells (K562, E:T ratio of 10:1) and rhIL-2 (200 IU/ml). On day 6, NK cells were transferred onto plates coated with the activating KLRG1 Ab or an isotype-control Ab. Cumulative data are shown as fold change compared with shCtrl-transduced cells stimulated with an isotype-control Ab. Events were gated on GFP+ (effectively transduced) cells only, and data are pooled from three independent experiments. Bars represent median ± interquartile range. The p values were calculated using the Student t test. (B) Population doubling time (PD) of shAMPK-transduced (red lines) and shCtrl-transduced (blue lines) NK cells stimulated with an activating KLRG1 Ab or an isotype-control Ab. Arrows represent times of stimulation with feeder cells and fresh rhIL-2. (C) Ki-67 expression on day 9 of cell culture. Analysis of telomerase reverse transcriptase (TERT) expression on day 9 from one representative experiment (D) and pooled data from three independent experiments (E). (F) DNA damage, as assessed by H2Ax phosphorylation (representative data from two independent experiments). (G) Apoptosis assessed by Annexin V staining on day 9 of cell culture, as described above. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. ns, p > 0.05.
KLRG1-induced inhibition of NK cell function is AMPK dependent. (A) Cytotoxic activity assessed by calcein-release lysis assay in purified KLRG1bright NK cells transfected with siRNA specific for KLRG1 (siKLRG1) or control siRNA (siCtrl) for 36 h and tested vis-à-vis the E-cadherin–expressing tumor cell line MCF-10A (4 h coculture). Throughout the cell culture, NK cells were kept in complete medium supplemented with 500 IU/ml rhIL-2. Release of granzyme B and IFN-γ under the same conditions are also shown. Reliance of KLRG1 on AMPK activity was probed by addition of the AMPK agonist A-769662 (150 μM) to the transfected cells 2 h prior to the cytotoxicity assay. A DMSO solution was used as vehicle control. Data are pooled from six donors (specific lysis) or five donors (granzyme B and IFN-γ). (B–G) KLRG1 ligation negatively regulates NK cell activity in AMPK-proficient cells only. Purified KLRG1bright NK cells were transduced with lentiviral vectors containing silencing shAMPK or shCtrl, as described. On days 6 and 9, NK cells were restimulated with feeder cells (K562, E:T ratio of 10:1) and rhIL-2 (200 IU/ml). On day 6, NK cells were transferred onto plates coated with the activating KLRG1 Ab or an isotype-control Ab. Cumulative data are shown as fold change compared with shCtrl-transduced cells stimulated with an isotype-control Ab. Events were gated on GFP+ (effectively transduced) cells only, and data are pooled from three independent experiments. Bars represent median ± interquartile range. The p values were calculated using the Student t test. (B) Population doubling time (PD) of shAMPK-transduced (red lines) and shCtrl-transduced (blue lines) NK cells stimulated with an activating KLRG1 Ab or an isotype-control Ab. Arrows represent times of stimulation with feeder cells and fresh rhIL-2. (C) Ki-67 expression on day 9 of cell culture. Analysis of telomerase reverse transcriptase (TERT) expression on day 9 from one representative experiment (D) and pooled data from three independent experiments (E). (F) DNA damage, as assessed by H2Ax phosphorylation (representative data from two independent experiments). (G) Apoptosis assessed by Annexin V staining on day 9 of cell culture, as described above. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001. ns, p > 0.05.
To further dissect the link between AMPK and KLRG1 function, we silenced AMPK in primary human NK cells, followed by KLRG1 ligation. We activated purified KLRG1bright NK cells with the MHC class I–deficient NK target cell line K562 and rhIL-2 for 48 h and then transduced cells with lentiviral vectors encoding short hairpin RNA to AMPK (shAMPK) or an irrelevant scrambled short hairpin RNA (control RNA; shCtrl). Four days posttransduction, we transferred NK cells to plates coated with KLRG1 or an isotype-control Ab and reactivated them up to 6 d, as above (for experimental design and transfection efficiency see Supplemental Fig. 4). The knockdown of AMPK restored NK cell expansion (Fig. 5B), proliferation (Fig. 5C), and hTERT expression (Fig. 5D, 5E) but reduced DNA damage (Fig. 5F) and apoptosis (Fig. 5G) compared with KLRG1bright NK cells transduced with shCtrl. For each function examined, the inhibitory effect induced by KLRG1 ligation was robust in NK cells transduced with shCtrl but was totally abolished in AMPK-silenced cells (Fig. 5B–G). Of note, AMPK knockdown did not alter KLRG1 cell surface expression in NK cells (data not shown). Thus, AMPK activation is a new inhibitory pathway of KLRG1 function in primary human NK cells, and blocking this pathway restores NK cell responsiveness.
Discussion
We identified a previously unrecognized function of the inhibitory receptor KLRG1 in linking cell surface inhibitory receptor signaling to AMPK activation in human NK cells. NK cells integrate signals from inhibitory and activating receptors to detect and eliminate infected and transformed cells (45, 46). Although activating NK cell receptors recognize stress-induced ligands on target cells, inhibitory NK cell receptors recognize MHC class I and MHC class I–like molecules on healthy cells (8, 46) as a means of protecting healthy tissue from NK cell attack. It is conceivable that an imbalance toward inhibitory immune-receptor signaling, as we now demonstrate for KLRG1 in individuals >70 y, increases the activation threshold of NK cells and leads to attenuation of effector function. Although this mechanism potentially protects from autoreactivity and collateral damage during infection, it might render hosts more susceptible to infection and/or malignancy by attenuating NK cell function. Therefore, we suggest that blocking of the KLRG1/AMPK signaling axis may boost NK cell activity in the older population.
Our data support a model whereby persistent (endogenous) AMPK activity is regulated through protection from dephosphorylation rather than by upstream kinase activation. The phosphatase PP2C is known to dephosphorylate (and thus inactivate) AMPK in vitro and in vivo (41, 42), but the precise physiological mechanism by which PP2C itself is regulated in humans was not known. We now show that KLRG1 internalizes upon ligation, binds directly to AMPK, and stimulates its function by inhibiting PP2C-like phosphatase activity. Correspondingly, KLRG1 ligation strongly diminished PP2C binding to AMPK in human NK cells. This links AMPK activation with surface inhibitory receptor signaling, suggesting that immune-inhibitory receptors, which increase with age, chronic viral infections, and cancer (14, 47, 48), may orchestrate AMPK-dependent metabolic pathways to inhibit NK cell function.
We further demonstrate that KLRG1 requires pre-existing AMPK activity to stimulate the AMPK pathway. Thus, KLRG1 seems to function as a natural AMPK enhancer in NK cells that is incapable of triggering de novo kinase activation but induces potent signal amplification. This occurs endogenously in highly differentiated NK cells that exhibit senescence characteristics, such as pre-existing DNA damage and spontaneous AMPK activity (32). We showed recently that endogenous DDR signaling activates AMPK in senescent T cells, and we now extend this observation to highly differentiated human NK cells. This raises the paradox of why constitutive activation of a low-energy sensor molecule would require continuous energy consumption to fuel its own activation in senescent cells. Because KLRG1 impedes AMPK dephosphorylation, we propose a model in which endogenous AMPK activity is maintained by active surface inhibitory receptor signaling rather than continuous de novo upstream activation. Beyond biochemical considerations, the central implication for this is that endogenous AMPK activity may be controlled by means of surface inhibitory receptors in human NK cells, a mechanism that was previously unknown.
It remains to be determined whether KLRG1 has a similar role in highly differentiated human T cells that also express high levels of KLRG1 (19, 49) and spontaneously activate AMPK (32). Furthermore, it is not known whether this pathway is shared with other T cell inhibitory receptors, such as CTLA-4 and PD-1, for which blocking Abs have been approved or are in clinical development.
In summary, we demonstrate that there is an intimate relationship between senescence and energy-sensing pathways in NK cells that can be further modulated by inhibitory receptor engagement. Intervention in this signaling axis may enhance NK cell activity.
Footnotes
This work was supported by the Swiss National Foundation (Grants P300PB_161092 and P2BSP3_151877 to B.M.-D.), the Wellcome Trust (Grant AZR00630 to A.L.), the Ministry of Education of Brazil (Grant BEX9414/14-2 to L.P.C.), and the Biotechnology and Biological Science Research Council (Grant BB/L005328/1 to A.N.A.). A.L. is a Sir Henry Wellcome Trust Fellow sponsored by Prof. Michael L. Dustin (University of Oxford). S.M.H. is funded by the William Harvey Research Foundation.
The online version of this article contains supplemental material.
Abbreviations used in this article:
- AMPK
AMP-responsive protein kinase
- BDS
Bright Detail Similarity
- hTERT
human telomerase reverse transcriptase
- KLRG1
killer cell lectin-like receptor G1
- MFI
mean fluorescence intensity
- PP2C
protein phosphatase 2C
- rh
recombinant human
- shAMPK
short hairpin RNA to AMPK
- shCtrl
short hairpin RNA (control RNA)
- siRNA
small interfering RNA.
References
Disclosures
The authors have no financial conflicts of interest.