Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Feb 13;57(2):287-302.e12.
doi: 10.1016/j.immuni.2024.01.011.

Signaling via a CD27-TRAF2-SHP-1 axis during naive T cell activation promotes memory-associated gene regulatory networks

Affiliations

Signaling via a CD27-TRAF2-SHP-1 axis during naive T cell activation promotes memory-associated gene regulatory networks

Carla A Jaeger-Ruckstuhl et al. Immunity. .

Abstract

The interaction of the tumor necrosis factor receptor (TNFR) family member CD27 on naive CD8+ T (Tn) cells with homotrimeric CD70 on antigen-presenting cells (APCs) is necessary for T cell memory fate determination. Here, we examined CD27 signaling during Tn cell activation and differentiation. In conjunction with T cell receptor (TCR) stimulation, ligation of CD27 by a synthetic trimeric CD70 ligand triggered CD27 internalization and degradation, suggesting active regulation of this signaling axis. Internalized CD27 recruited the signaling adaptor TRAF2 and the phosphatase SHP-1, thereby modulating TCR and CD28 signals. CD27-mediated modulation of TCR signals promoted transcription factor circuits that induced memory rather than effector associated gene programs, which are induced by CD28 costimulation. CD27-costimulated chimeric antigen receptor (CAR)-engineered T cells exhibited improved tumor control compared with CD28-costimulated CAR-T cells. Thus, CD27 signaling during Tn cell activation promotes memory properties with relevance to T cell immunotherapy.

Keywords: CAR-T cell therapy; CD27; CD8(+) T cell; SHP-1 phosphatase; chimeric antigen receptor; costimulation; memory and effector fate determination; naive T cell activation.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests C.A.J.-R., C.E.C., and S.R.R. are inventors on a patent (“engineered trimeric CD70 proteins and uses thereof”; WO2021072127A3) filed by Fred Hutchinson Cancer Center and licensed by Lyell Immunopharma. S.R.R. was a founder, has served as an advisor, and has patents licensed to Juno Therapeutics; S.R.R is a founder of and holds equity in Lyell Immunopharma and has served on the advisory boards for Adaptive Biotechnologies and Nohla.

Figures

Figure 1 |
Figure 1 |. CD70 ligand binding induces rapid surface CD27 receptor down-modulation.
(A) Model of the Fc-fused dimer of assembled CD70 single chain trimers. (B) Representative histograms of a CFSE-dilution assay of non-activated (n.a.) or 72 h activated cells following stimulation with (left) αCD3 and CD70DT or (right) combinations of αCD3, αCD28 and CD70DT [αCD3|αCD28|CD70DT; μg/mL]. (C) Western blot of lysates from CD8+ Tn that were n.a. or activated with the indicated concentrations of αCD3+αCD28+CD70DT for 5 min, 15 min, 24 h and 72 h. Blots for CD27 (full-length 55kDa and cleaved 28–32 kDa) and beta 2 microglobulin (B2M) loading control are shown for each stimulation condition. (D) (Left) Representative histogram and (right) quantification of CD27 surface expression on n.a. and activated CD8+ Tn (n=4 donors). (E) Representative histogram showing down-modulation of CD27 surface expression induced by CD70DT requires αCD3 stimulation. (F) Illustration of K32 APC and T cell activation. (G) (Left) Representative histogram and (right) quantification of CD27 surface expression on CD8+ Tn co-cultured [1:1] with αCD3-coated (5 μg/mL) K32 or CD70-transgene expressing K32 cells (n=3 donors). (H) Illustration of BM derived Ova-presenting DC and OT1 T cell activation. (I) Representative histogram showing CD27 surface expression on murine OT-1 CD8+ Tn co-cultured [2:1] with activated murine BM derived OVA-presenting DCs in the presence of IgG or αCD70-blocking antibody (FR70). Representative experiment is shown. Data are shown as mean ± s.e.m. and analyzed by one-way ANOVA (D,G). Illustrations (C,F,H) created using BioRender. See also Figure S1.
Figure 2 |
Figure 2 |. CD27 surface expression is regulated by clathrin-mediated endocytosis.
(A) Illustration of clathrin-mediated CD27 receptor endocytosis (CME). (B) (Left) Representative histogram and (right) quantification of CD27 surface expression of non-activated (n.a.) and activated (2–15 min) CD8+ Tn treated ±CME inhibitors (CMEi) ES9–17 (100 μM), Mdivi-1 (50 μM) or a combination of both (right: n=5 donors). (C) (Left) Representative histogram of fixed and permeabilized T cells and (right) quantification of total cellular CD27 in n.a. or 10 min activated CD8+ Tn treated ±CMEi, autophagy inhibitor (ULK-101; 5 μM) or proteasome inhibitor (Lactacystin; 20 μM) (n=5–28 donors). Data are shown as mean ± s.e.m. and analyzed by one-way ANOVA (B) or paired two-tailed Student’s t-test (C). Illustration (A) created using BioRender. See also Figure S2.
Figure 3 |
Figure 3 |. CD27-TRAF2-SHP-1 modulates Lck and downstream signaling induced by αCD3+αCD28.
(A) TRAF2 immunoprecipitation of lysates from non-activated (n.a.) and 10 min activated CD8+ Tn at indicated doses of αCD3, αCD28 and CD70DT [αCD3|αCD28|CD70DT; μg/mL]. Immunoblots for TRAF2, CD27, B2M are shown. (B) TRAF2 immunoprecipitation of lysates from n.a. and CD8+ Tn activated for 10 min under the indicated conditions and treated with Latrunculin A (LatA, 0.05 μM) or DMSO. Immunoblots for TRAF2, CD27, GAPDH are shown (1 representative donor of n=3). (C) TRAF2 immunoprecipitation of lysates from n.a. and CD8+ Tn activated for 10 min under the indicated conditions. Immunoblots for TRAF2, CD27, SHP-1, B2M are shown (n=1). (D) Representative immunoblots of lysates from n.a. and CD8+ Tn activated for 10 min under the indicated conditions for phospho-SHP-1(Y564) and B2M. Fold-change of band intensity over B2M loading control was calculated (n=4 donors). (E) SHP-1 immunoprecipitation of lysates from n.a. and 10 min activated CD8+ Tn. Immunoblots for SHP-1, phospho-SHP-1(Y564), Lck, GAPDH are shown (1 representative donor of n=2). (F) Representative immunoblots of lysates from n.a. and 10 min activated CD8+ Tn for Lck, phospho-Lck (Y394, Y505) and B2M. Fold-change of band intensity over B2M loading control was calculated (n=8–12 donors for Y394 and n=4–6 donors for Y505). (G) Immunoblots of lysates from n.a. and 10 min activated CD8+ Tn treated with SHP-1 inhibitor (TPI-1, 0.4 μM) or DMSO for total Lck, phospho-Lck(Y394), and B2M. Fold-change of band intensity over B2M loading control was calculated (n=5 donors). (H,I) Frequency of phosphor-ERK1/2 (T202, Y204) positive T cells in n.a. and 15 min activated CD8+ Tn assessed by phosphor-flow (H: n=14 donors, I: n=13 donors). (J) (Left) Representative histogram and (right) quantification of phospho-ERK1/2 (T202, Y204) in n.a. and 15 min CD8+ Tn treated with DMSO or TPI-1 (0.4 μM) by phospho-flow (n=8 donors). (K) Illustration of CRISPR-Cas9-based SHP-1 knock-out in human CD8+ Tn cells and culture conditions. (L) Flow cytometry-based assessment of (left) SHP-1 expression in wildtype (WT, blue) or SHP-1 deficient (KO, red) CD8+ Tn cells and (right) frequency of phospho-ERK1/2 (T202, Y204) positive T cells in n.a. and WT and KO CD8+ Tn activated for 15 min under the indicated conditions (1 representative donor of n=2). All data are shown as mean ± s.e.m. and were analyzed by paired two-tailed Student’s t-test (D,F-J). Illustrations in (K) created using BioRender. See also Figure S3.
Figure 4 |
Figure 4 |. CD27 costimulation promotes memory characteristics.
CD8+ Tn are activated under various conditions of plate-bound [αCD3|αCD28|CD70DT; μg/mL] or αCD3+αCD28 beads [at 3:1 bead:cell ratio] for 72 h and expanded for 9 days. (A) Cell cycle analysis was performed at 48 h and 96 h using 7-AAD nucleic acid stain to determine the fraction of cells in G0+G1, S, G2+M (n=3–10 donors). (B) Fold-expansion over input of activated CD8+ Tn at respective timepoints (n=3–10 donors). (C) (Left) Representative contour plots showing CD27 and TCF1 expression in CD8+ T cells 9 days after stimulation and (right) quantified fraction of CD27+TCF1+ cells (n=5–10 donors, 2 independent experiments). (D) MFI of CD127 and CCR7 surface expression 9 days after stimulation (n=5 donors). (E) Percent expression of CD45RA and CD45RO on T cells expanded for 9 days is shown (n=4–11 donor of 3 independent experiments). (F) [13C] glucose uptake from media by CD8+ Tn at various timepoints after stimulation (n=3 donors). Ion counts normalized to timepoint 0 h. (G) Seahorse-based glucose stress test (left) assessing changes in extracellular acidification (ECAR) and (right) measuring glycolytic capacity of CD8+ T cells 5 days after stimulation (1 representative donor à 10 replicate wells). Glucose, oligomycin (oligo) and 2-Deoxy-D-glucose (2-DG) were added as indicated. (H) Seahorse-based mitochondrial stress test (left) assessing changes in oxygen consumption rate (OCR) and (right) measuring spare respiratory capacity of CD8+ T 10 days after stimulation (1 representative donor à 10 replicate wells). Oligomycin (oligo), phenylhydrazone (FCCP) and rotenone and antimycine A (Rot+AA) were added as indicated. All data are shown as mean ± s.e.m. and were analyzed by two-way ANOVA (A,B,F), paired (C,D,E) and unpaired (G,H) two-tailed Student’s t-test. Illustration in (A) created using BioRender. See also Figure S4.
Figure 5 |
Figure 5 |. CD27 costimulation induces distinct gene regulatory circuits early after T cell activation.
Single cell multiome-seq of CD8+ Tn that are non-activated (n.a.) and activated for 24 h with αCD3+αCD28 beads [at 3:1 bead:cell ratio] or plate-coated αCD3+αCD28+CD70DT ([5|0|5]; μg/mL) (n=1 donor). (A) Subclustered UMAP and (B) top differentially expressed genes in respective subclusters of single cell RNA-seq data. (C-F) ATAC-seq of the 3 main clusters showing (C-E) coverage plots of FOXP1, STAT1 and IFNG gene loci and linked (red=repressive, blue=activating) promoter regulatory gene peaks. (F) ATAC-footprints of TBX21 and NFATC2 transcription factor motifs. (G,H) Combined RNA-seq and ATAC-seq showing (G) heatmap-based correlation of RNA expression to DNA accessibility of indicated top-regulated transcription factors and (H) inferred target gene regulation of selected transcription factors. See also Figures S5 and S6.
Figure 6 |
Figure 6 |. CD27 costimulation of CAR-T cells during manufacturing improves in vivo function.
(A) Murine B cell aplasia model. Murine CD45.1+CD8+ T cells were isolated from spleen, activated with plate-coated αCD3 [1 μg/mL] and αCD28 or CD70DT [each at 1 μg/mL], transduced with a retrovirus encoding a mCD19_4-1BB_CD3ζ CAR and EGFRt (transduction marker), and transferred at a cell dose of 1×106 into lymphodepleted (Cytoxan) CD45.2+ C57BL/6 mice. (B) Quantification of EGFRt+CD8+ CAR-T cells in peripheral blood of mice (n=12–13, 2 pooled experiments). (C) Monitoring of CD19+ B cells in the peripheral blood of CAR-T cell infused or control mice (B cell reconstitution cut-off >= 1%). (D) Quantification of EGFRt+CD8+ CAR-T cells in peripheral blood of mice 71 days post infusion (n=5 mice). (E-G) (E) TCF1 and (F,G) Ki67 expression in EGFRt+CD45.1+ or non-transduced (nTd) EGFRtCD45.1+ T cells in BM and spleen harvested at day 4 (E,F) and BM harvested at day 21 (G) after adoptive transfer (AdTf) (n=4–5 mice per timepoint and group). (H) Raji-lymphoma xenograft model. Human bulk CD8+ T cells were isolated from peripheral blood of healthy donors, activated with plate-coated αCD3+αCD28+CD70DT ([5|0|5]; μg/mL) or αCD3+αCD28 beads [at a 3:1 bead:cell ratio] prior to transduction with a lentivirus encoding a hCD19_4–1BB_CD3ζ CAR and EGFRt. T cells were transferred at a dose of 0.7×105 to tumor bearing mice. (I) Frequency of EGFRt+CD8+ CAR-T cells per total human CD45+ T cells in peripheral blood of mice. (J) Cell surface phenotype of peripheral blood EGFRt+CD8+ CAR-T cells 7 days post infusion. (K) Bioluminescence imaging of ffluc+ Raji cells in control (untreated) or CAR-T cell treated mice. (I-K) n=9 mice per group. All data are shown as mean ± s.e.m. and were analyzed by Log-Rank (Mantle-Cox) test (C), Turkey’s multiple comparison test (B,I), unpaired two-tailed Student’s t-test (D-G,J) and two-way ANOVA (K). Illustrations in (A,H) created using BioRender. See also Figure S7 & Figure S8.

References

    1. Wiesmann A, Phillips RL, Mojica M, Pierce LJ, Searles AE, Spangrude GJ, and Lemischka I (2000). Expression of CD27 on murine hematopoietic stem and progenitor cells. Immunity 12, 193–199. 10.1016/s1074-7613(00)80172-7. - DOI - PubMed
    1. Hintzen RQ, de Jong R, Lens SM, Brouwer M, Baars P, and van Lier RA (1993). Regulation of CD27 expression on subsets of mature T-lymphocytes. J Immunol 151, 2426–2435. - PubMed
    1. van Lier RA, Borst J, Vroom TM, Klein H, Van Mourik P, Zeijlemaker WP, and Melief CJ (1987). Tissue distribution and biochemical and functional properties of Tp55 (CD27), a novel T cell differentiation antigen. J Immunol 139, 1589–1596. - PubMed
    1. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, and van Lier RA (1997). Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med 186, 1407–1418. 10.1084/jem.186.9.1407. - DOI - PMC - PubMed
    1. Gattinoni L, Speiser DE, Lichterfeld M, and Bonini C (2017). T memory stem cells in health and disease. Nat Med 23, 18–27. 10.1038/nm.4241. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances