Summary:
The interaction of the tumor necrosis factor receptor (TNFR) family member CD27 on naïve 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 the effector associated gene programs, which are induced by CD28 costimulation. CD27-costimulated chimeric antigen receptor (CAR) engineered T cells exhibited improved tumor control compared to CD28-costimulated CAR-T cells. Thus, CD27 signaling during Tn cell activation promotes memory properties with relevance to T cell immunotherapy.
Graphical Abstract

eTOC blurb:
Interaction of CD27 on naïve T cells with CD70 on antigen presenting cells is necessary for T cell memory fate determination. Jaeger-Ruckstuhl et al. engineer a trimeric CD70-ligand and assess consequences on CD27 ligation on receptor regulation, early proximal signaling, transcriptional and epigenetic states, revealing how modulation of T cell activation by CD27 affects fate commitment and efficacy of T cell immunotherapy.
Introduction:
The expression of the tumor necrosis factor receptor (TNFR) family member CD27 is confined to the earliest hematopoietic cell progenitors and to cells of the lymphoid cell lineage 1–3. CD27 is highly expressed by naïve CD8+ T cells (Tn), maintained in stem-cell (Tscm) and central memory (Tcm) cells, and expressed at lower levels in only a subset of effector memory (Tem) and terminal effector (Teff) cells 4,5. During antigen priming of Tn cells, the interaction of CD27 with its homotrimeric ligand CD70, which is transiently induced on antigen presenting cells (APCs), is necessary for T cell memory fate determination 6–9. Overexpression of CD27 in HEK 293 cells induces pro-survival signals via Nuclear Factor κB (NFκB)-inducing kinase (NIK), stress-activated protein kinase (SAPK) and c-JUN N-terminal kinase (JNK) 10. In murine T cells, TNFR Associated Factor 2 (TRAF2)- and Nck-interacting kinase (TNIK) act downstream of the CD27 receptor to promote Wnt pathway activation 11. These findings identify important CD27 functions but do not fully explain how CD27 might cooperate with the T cell receptor (TCR) and CD28 signaling to affect T cell fate.
The potential to enrich and preserve memory T cell states during in vitro T cell expansion is of interest for adoptive T cell therapy 12. T cells engineered to express chimeric antigen receptors (CAR) that target tumor-associated antigens exhibit potent activity in hematologic malignancies 13. Preparation of CAR-T cells for adoptive transfer involves activation with αCD3 and αCD28 antibodies to facilitate gene insertion and increase cell numbers but leads to progressive T cell differentiation that may compromise long-term efficacy. Higher numbers of CD27+ cells in the starting T cell population and CAR-T products containing gene expression signatures of less differentiated cells provide for more durable antitumor responses 14,15. Given the role of CD27 in promoting T cell memory in vivo, we sought to understand the regulation and function of CD27 after ligation with CD70 in vitro. To engage CD27 as a trimer we designed functional CD70 trimeric ligands 16. We found that CD27 ligation by plate-coated CD70 in conjunction with TCR stimulation provided by αCD3 monoclonal antibody (mAb) promoted rapid internalization of CD27 in a dose-dependent manner. This effect preceded CD27 cleavage and was observed in antigen-specific CD8+ T cells interacting with APC expressing physiologic levels of CD70. CD27 costimulation improved proliferation of Tn cells activated with αCD3 alone, but when strongly engaged antagonized CD28 signaling, an effect mediated by co-recruitment of TRAF2 and the phosphatase SHP-1, and dephosphorylation of the tyrosine kinase Lck. Compared to CD28 costimulation of αCD3-activated Tn cells, costimulation via CD27 transiently restrained cell-cycle entry and induced a distinct metabolic, transcriptional and epigenetic state. Consequently, CD27-costimulated CAR-T cells exhibited a phenotype similar to memory T cells, and had improved therapeutic activity in models of chronic antigen stimulation and cancer.
Results
Binding of CD27 by CD70 induces receptor endocytosis
To investigate the consequences of CD27 ligation, we designed an Fc-fused dimer of single-chain CD70 trimers (CD70DT) that provides costimulation to T cells activated with αCD3 mAb or with a combination of αCD3 and αCD28 mAb (Figures 1A, 1B, S1A). CD70DT alone did not induce CD69 expression or cell division as measured by CFSE dye dilution indicating dependence on αCD3 engagement for costimulatory effects (Figure S1B). To determine how ligand engagement might affect CD27 expression, we used western blot with an antibody specific to the CD27 endodomain to evaluate total CD27 protein levels in lysates from Tn cells stimulated with αCD3 mAb alone, with αCD3 combined with αCD28 mAb, or with αCD3 combined with CD70DT. Consistent with previous reports that CD27 undergoes cleavage in activated T cells 17,18, a 28 kD cleavage product was detected in all stimulation conditions at 24 h and was prominent at 72 h (Figure 1C). As early as 5 min after Tn activation we observed a marked reduction of the intact 55 kDa CD27 single chain that was dependent on ligation by CD70 and occurred before the 28 kDa cleavage product was detected (Figure 1C). Assessment of cell surface CD27 by flow cytometry revealed a rapid dose-dependent reduction after ligation with CD70DT in αCD3 and αCD3+αCD28-stimulated T cells but minimal loss with CD70DT ligation alone (Figures 1D, S1C, 1E). The rapid decline in cell surface CD27 after αCD3+CD70DT stimulation was not associated with differences in CD27 mRNA levels, prevented by inhibiting matrix metalloproteinases responsible for cleavage, or due to competition for αCD27 antibody binding-epitopes by CD70DT (Figures S1D–S1F). CD27 internalization occurred in all CD8+ T cell subsets (Tn, Tcm, Tem, CD45RA+ Tem (Temra)) activated with αCD3+CD70DT (Figure S1G).
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.
To determine if CD27 internalization was also observed upon interaction with CD70 expressing APCs, we co-cultured Tn cells with K562-derived APCs that stably expressed the Fcγ receptor CD32 (K32) 19 and were transduced with a vector encoding CD70 (Figures 1F, S1H). CD27 internalization was observed in co-cultures with CD70-transgene expressing K32 cells presenting αCD3 mAb, but not with K32 cells that did not express CD70 (Figure 1G). Rapid CD27 receptor internalization was also observed in murine OT-1 T cells co-cultured with matured bone marrow (BM) derived ovalbumin transgenic dendritic cells (DCs) that expressed endogenous levels of CD70 and was inhibited by the addition of a CD70 blocking antibody to prevent CD27 binding (Figures 1H and 1I).
We next sought to define the mechanism for the rapid loss of cell surface CD27 after ligand binding. TNFR1 is internalized by clathrin-mediated endocytosis (CME) upon TNF binding suggesting CD27 may undergo similar regulation 20,21. CME involves clathrin, dynamin, actin and endophilin for the initiation of membrane pit formation and subsequent endocytic vesicle scission 22. We therefore measured CD27 surface expression on αCD3+CD70DT-activated Tn cells in the presence or absence of clathrin-heavy chain inhibitor (ES9–17), dynamin-related protein inhibitors (Mdivi-1, Dynasore, Dyngo-4a) or combinations thereof (Figure 2A). CD27 surface expression was substantially increased in the presence of each CME inhibitors in both the plate-coated and K32 APC activation systems, supporting CME as the mechanism for rapid CD27 internalization (Figures 2B, S2A, S2B). Inhibitors of autophagy initiation (ULK-101) and proteasomal degradation (Lactacystin) also partially rescued total cellular CD27 levels providing evidence for lysosomal and proteasomal degradation of internalized CD27 (Figure 2C). Levels of cell surface CD27 were gradually restored over 7 days in culture (Figure S2C). These data identify a mechanism by which CD27 surface expression on activated T cells is regulated by CD70 binding through rapid ligand-induced CME followed by intracellular receptor degradation and re-expression.
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.
A CD27-TRAF2-SHP-1 axis regulates Lck and downstream signaling induced by TCR and CD28 ligation
CD27 propagates signals via association with TRAFs 23. To determine if TRAF2 was recruited during CD27 receptor internalization, we immunoprecipitated TRAF2 in lysates of non-activated or αCD3-stimulated Tn cells in the presence of various doses of αCD28 mAb or CD70DT and examined its association with CD27 by western blot. Whereas CD70DT binding increased TRAF2 association with CD27 in a dose-dependent manner, neither αCD3 alone nor αCD3+αCD28 stimulation promoted its association with CD27 (Figures 3A, S3A). The addition of actin polymerization inhibitor Latrunculin A (LatA) prevented CD27 endocytosis and reduced TRAF2 association with CD27 in Tn cells that were stimulated with high dose of CD70DT (Figure 3B, S3B).
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.
Irrespective of the T cell activation state, we found that TRAF2 interacted with SHP-1 but not SHP-2 (Figures 3C, S3C). This led us to hypothesize that CD27 engagement might alter SHP-1 activity. We therefore assessed SHP-1 phosphorylation at activating (Y536, Y564) and inactivating (S591) residues in unstimulated and 10 min stimulated Tn cells. No CD27 signaling-induced changes in phosphorylation were observed at residues SHP-1Y536 and SHP-1S591, however SHP-1Y564 phosphorylation was significantly increased when CD27 was ligated in Tn stimulated with αCD3+αCD28 (Figure S3D). SHP-1Y564 phosphorylation was minimally increased with αCD3+CD70DT stimulation (Figure 3D), however, the total cellular amount of THEMIS, a negative regulator of SHP-1 24, was reduced in Tn cells activated with αCD3+CD70DT and αCD3+αCD28+CD70DT (Figure S3E). This suggests that CD27 ligation acts to induce SHP-1 activity, most notably in Tn cells activated with αCD3+αCD28.
We then asked whether CD27 induced changes in SHP-1 might regulate phosphorylation of the SHP-1 substrate Lck, which is crucial for T cell activation 25. We first assessed whether SHP-1 interacted with Lck by immunoprecipitating SHP-1 from non-activated and 10 min stimulated Tn cells. SHP-1 co-immunoprecipitated with Lck in all αCD3-activated conditions, independent of SHP-1 phosphorylation state or whether CD28 or CD27 costimulation was provided (Figure 3E). When combined with αCD3+αCD28 activation, CD27 ligation resulted in de-phosphorylation of activating LckY394 but not inhibitory LckY505 residues as measured by immunoblot of whole cell lysates. CD27 ligation had only minimal effect on LckY394 in cells stimulated with αCD3 alone (Figure 3F). The addition of an IgG1-Fc control at the same concentration as CD70DT did not result in de-phosphorylation of LckY394, demonstrating the requirement for CD27 engagement via its ligand CD70 to modulate Lck (Figure S3F). When higher doses of αCD28 mAb were used LckY394 was also de-phosphorylated in the presence of CD70DT (Figure S3G), suggesting recruitment of TRAF2-SHP-1 modulates the sustained activity of Lck mediated by CD28 signaling of various strengths 26. Small molecule inhibition of SHP-1 prevented de-phosphorylation of LckY394 in αCD3+αCD28+CD70DT-activated Tn cells (Figure 3G). Collectively, this data shows that CD27 ligation results in assembly of a CD27-TRAF2-SHP-1 complex and promotes SHP-1 phosphatase-induced modulation of Lck activity in Tn cells activated through the TCR and CD28.
Lck activity contributes to the magnitude of the extracellular signal-regulated kinase 1/2 (ERK1/2) 27 and phosphatidylinositol 3-Kinase (PI3K) 28 response leading us to speculate that CD27-TRAF2-SHP-1 modulation of Lck might affect ERK1/2 and AKT Serine and Threonine Kinase 1 (AKT) signaling. αCD28 activation enhanced ERK1/2 and AKT phosphorylation over αCD3 activation alone in a dose-dependent manner. Increasing doses of CD70DT combined with αCD3 resulted in a minor increase in ERK1/2 phosphorylation that was significant only at the highest dose of CD70DT, and no increase in AKT phosphorylation was observed (Figures 3H, S3H). When added to αCD3+αCD28, CD70DT reduced phosphorylation of ERK1/2 and AKT significantly compared to αCD3+αCD28 alone (Figures 3I, S3I). The CD70-mediated modulation of ERK1/2 phosphorylation was also observed using the K32 cell-based co-culture model (Figure S3J). We evaluated ERK1/2 phosphorylation over a range of plate-coated αCD3, αCD28 and CD70DT concentrations and found that ERK1/2 was only modulated at higher CD70DT doses that induced CD27 internalization (Figures S3K, 1D, S1C). The reduction in ERK1/2 phosphorylation by CD70DT was partially prevented by addition of the SHP-1 inhibitor, further linking this effect of CD27 ligation to the TRAF2-SHP-1-Lck signaling axis (Figure 3J). Because small molecule inhibitors may have non-specific effects, we sought further evidence for the involvement of SHP-1 activity downstream of CD27-TRAF2 in regulating ERK1/2 phosphorylation. We generated a SHP-1 gene deletion in resting Tn cells using CRISPR-Cas9-based editing (Figure 3K). Under conditions that favor the maintenance of a CD45RA+CD62L+CCR7+ Tn cell phenotype, we achieved a SHP-1 gene deletion efficiency of up to 88 % after 7–8 days in culture, compared to control Tn cells cultured under identical conditions (Figures S3L, S3M). Analysis of ERK1/2 phosphorylation after activation of SHP-1 wildtype (WT) and SHP-1 deleted (KO) Tn cells demonstrated that SHP-1 was required for the modulatory effect on ERK1/2 phosphorylation mediated by CD27 costimulation (Figure 3L).
We next evaluated whether CD27 receptor endocytosis was required for the modulation of Lck and ERK1/2 phosphorylation by activating Tn cells in the presence or absence of LatA. Inhibition of CME using LatA inhibited CD27 receptor internalization (Figure S3B) and prevented dephosphorylation of Lck and the reduction in ERK1/2 phosphorylation (Figures S3N, S3O). Collectively, these data show that TCR and CD28 signaling is modulated by CD27 ligation and CME through SHP-1 phosphatase-mediated de-phosphorylation of Lck and reduction in downstream ERK1/2 and AKT signaling.
CD27 costimulation delays cell-cycle entry and promotes a memory phenotype
The strength of TCR and costimulatory signaling are determinants of cell proliferation rate, cell differentiation, and metabolic adaptation 29. As observed in Figure 1B, the addition of high CD70DT doses to αCD3-activated Tn cells enhanced cell proliferation over αCD3 alone, indicating that CD27 costimulation has effects on T cells that are independent of its modulatory role on CD28 signaling. To measure how CD27 engagement affects cell division, we assessed cell-cycle states at 48 h and 96 h after αCD3 stimulation alone or combined with CD27 or CD28 costimulation. In addition to plate-coated reagents, we included αCD3+αCD28 beads that are used to propagate T cells for adoptive T cell therapy. CD27 costimulation resulted in slightly more T cells in S and G2+M phase at 48 h post Tn cell activation compared to αCD3 activation alone, whereas only beads and the higher dose of plated-coated αCD28 mAb strongly promoted cell cycle entry (Figure 4A). Activation with αCD3+CD70DT transiently delayed cell-cycle entry compared with αCD3 activation, similar to the lower dose of αCD28-activated Tn cells (Figure 4A). Assessment of cell counts after 9 days of culture revealed that stimulation with αCD3+CD70DT resulted in a moderate T cell expansion, which was inferior to αCD3+αCD28 beads and strong αCD3+αCD28 activation, but superior to weak αCD3+αCD28 activation or αCD3 activation alone (Figure 4B).
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.
To examine how CD27 signaling might affect cell differentiation at the end of the 9 day expansion, we assessed expression of T cell factor 1 (TCF1), a transcription factor associated with T cell stemness 30. αCD3+CD70DT stimulation of Tn cells resulted in a higher fraction of TCF1+CD27+ expressing T cells than observed with all αCD3+αCD28 conditions or with αCD3 stimulation alone (Figure 4C). Only CD27 and strong CD28 costimulation resulted in a high expression of CD127 and CCR7 (Figure 4D) whereas T cells stimulated with αCD3 alone or with weak CD28 costimulation expressed these markers at lower levels, despite their similarly low expansion. Compared to αCD3+αCD28 bead-activated T cells, αCD3+CD70DT-activated Tn cells maintained a higher fraction of CD45RA positive and CD45RO negative cells, consistent with enrichment of T cells with a Tscm phenotype (Figure 4E). αCD3+CD70DT-activated T cells also exhibited a higher frequency of Interleukin 2 (IL2) positive cells and lower frequency of interferon gamma (IFNγ) positive cells after PMA and ionomycin stimulation (Figure S4A). Thus, CD27 costimulation alone promotes moderate cell proliferation and preserves a memory cell surface phenotype.
T cell differentiation state and cellular metabolism are closely linked 31. Extensive glycolysis is associated with adoption of an effector rather than memory fate 32 and coincides with activation-induced mitochondrial remodeling 33. To evaluate how CD27 costimulation affected glucose consumption we measured depletion of [13C] glucose in the culture supernatants after stimulation of Tn cells with αCD3+CD70DT compared to αCD3 or αCD3+αCD28 beads. We observed increased glucose depletion upon αCD3 activation that was greater when CD27 or CD28 costimulation was provided. At 5 h post Tn cell activation, the rate of glucose consumption in CD27-costimulated T cells slowed compared with CD28-costimulated T cells (Figure 4F). The lower glycolytic state after αCD3+CD70DT activation was maintained over 5 days of culture as assessed by reduced extracellular acidification compared to αCD3+αCD28 bead-activated T cells (Figure 4G). In order to address how the observed differences in expansion and effector glycolytic metabolism affected the mitochondrial compartment, we assessed mitochondrial mass using MitoTrackergreen. The mitochondrial mass of αCD3+CD70DT-activated T cells was significantly lower compared to αCD3+αCD28 bead-activated cells at the end of a 9-day culture (Figure S4B). Although αCD3+CD70DT-activated cells harbored a reduced mitochondrial mass, these cells possessed a higher basal oxygen consumption rate and improved spare respiratory capacity compared to αCD3+αCD28 bead-activated Tn cells (Figures 4H, S4C). Taken together, our data shows that CD27 costimulation transiently delays cell-cycle entry and promotes the acquisition of memory cell surface phenotype, functional, and metabolic properties, independent of its modulatory effect on CD28.
CD27 costimulation induces distinct gene regulatory networks
To better understand differences in the transcriptional and epigenetic responses induced by CD27 or CD28 costimulation, we performed multi-ome single cell sequencing of Tn cells before and 24 h post stimulation with αCD3+CD70DT or αCD3+αCD28, a time point when T cells had not yet undergone their first cell division. Dimensionality reduction of transcriptomic data identified an intermediate cell population emerging from the naïve cluster and bifurcating into two major branches delineated by CD28 or CD27 costimulation. The CD28 branch was further divided into sub-clusters discriminated by high and low IL2 transcript levels, whereas the CD27 branch was subclustered by differential expression of effector differentiation repressor Helios encoded by IKAROS Family Zinc Finger 2 (IKZF2) 34 (Figure 5A). Differential gene expression analysis revealed high transcript levels of the regulatory transcription factors (TF) Forkhead Box (FOX) Protein P1 (FOXP1) 35,36, stemness-associated TF TCF7 37, inhibitor of glycolysis Thioredoxin Interacting Protein (TXNIP) 38 and epigenetic repressor Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) 39 in resting Tn cells. Tn cells that were αCD3+CD70DT-activated for 24 h maintained high transcript levels of these Tn-associated genes, which were strongly decreased in response to CD28 costimulation. Genes that were induced in response to CD27 costimulation included the NF-κB cofactor 40,41 and quiescence regulator 42 Signal Transducer And Activator Of Transcription 1 (STAT1), protein arginine methyltransferase 2 (PRMT2), transcriptional repressor Zinc Finger And BTB Domain Containing 20 (ZBTB20), costimulatory receptor CD96 43, nuclear TCF1-cofactor Lymphoid Enhancer Binding Factor 1 (LEF1) as well as memory-associated surface markers SELL and CCR7. Unique CD28 costimulation-induced genes were associated with cell division [Cyclin D2 (CCND2), Tubulin Beta Class I (TUBB), Marker of Proliferation Ki-67 (MKI67)], effector metabolism [Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH), Enolase 1 (ENO1), Fatty Acid Binding Protein 5 (FABP5), MYC], stress response [ Heat Shock Protein 90 (HSP90)] and IL2 sensitivity [IL2, IL2 receptor alpha (IL2RA)] (Figures 5B, S5A).
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.
ATAC-seq analysis revealed that the transition from resting Tn cells to 24 h αCD3+αCD28-activated cells induced extensive remodeling of the DNA architecture, broadly enhancing its accessibility. The transition from resting Tn cells to 24 h αCD3+CD70DT-activated cells resulted only in a modest increase in DNA accessibility, suggesting differential access to TF networks (Figure S5B). The CD28 costimulation induced decrease of FOXP1 and TCF7 expression detected 24 h after activation resulted from increased DNA accessibility of promoter repressive regulatory elements (Figures 5C, S5C). CD27 costimulation increased STAT1, TCF7 and CCR7 expression, and was associated with increased DNA accessibility of promoter enhancing gene regions (Figures 5C, 5D, S5D). CD28 but not CD27 costimulation broadly increased accessibility at IFNG and IL2 promoter enhancing gene regions, two core transcriptional features of effector T cell differentiation 44 (Figures 5E, S5E). In line with a progressive acquisition of effector functions, αCD3+αCD28-stimulated T cells revealed distinct T-Box Transcription Factor 21 (TBX21), Nuclear Factor of Activated T cells 2 (NFATC2), FOS and JUN motif footprints, which were completely absent or attenuated in αCD3+CD70DT-activated cells (Figures 5F, S5F).
By integrating transcriptomic and motif accessibility data we were able to identify putative TF regulators and target genes in resting Tn cells and 24 h activated Tn cells (Figure S5G). We observed that αCD3+αCD28 activation resulted in overall broad TF motif accessibility that was only correlated with transcript levels in a subset of candidates (Figure S5H). In order to infer highly utilized TFs, we correlated transcript levels with motif accessibility. A particularly high positive correlation score was obtained in naïve and 24 h αCD3+CD70DT-activated cells for TF regulatory element Activating Transcription Factor 2 (ATF2) 45, the T cell quiescence regulator FOXP1 35,36, and the FOX Protein TFs J3 (FOXJ3) and O1 (FOXO1), both occupying an enhancer located upstream of the TCF1 locus and implicated in memory formation 46,47. However, 24 h αCD3+αCD28-activated cells showed strong positive correlation for TBX21, MYC-Associated Zinc Finger Protein (MAZ) and NFATC2, indicative of enhanced effector programming (Figure 5G). Lastly, we looked at inferred TF target genes in resting Tn cells and 24 h activated Tn cells to confirm gene regulatory network specificity. Analysis showed that the ATF2 network was induced in 24 h αCD3+CD70DT-activated cells when compared to naïve cells. Consistent with the low transcript-to-motif correlation (Figure 5G), interference-based ATF2 utilization was reduced in response to 24 h αCD3+αCD28 activation. Further, we observed particularly high utilization of quiescence- and stemness-associated TFs including FOXJ3, FOXP1, FOXO1, TCF7 and LEF1 in Tn cells. As expected, αCD3+αCD28 24 h-activated T cells showed significantly reduced utilization of these circuits. However, CD27 costimulation, but not CD28, preserved a stemness-like DNA architecture, limiting the transition towards effector differentiation. Consistent with acquisition of an activated T cell state, both CD27- and CD28-costimulated cells showed induction of STAT1 circuitry. Reflective of an effector differentiation state, CD28 costimulation strongly engaged TBX21, MAZ and NFATC2 and enhanced target gene expression (Figure 5H). In summary, this data suggests that the epigenetic alterations evoked in response to CD27 costimulation enhances regulatory TF circuits that preserve a stem-like T cell state and hinder effector differentiation.
Given the observed modulatory role of CD27 costimulation when provided in combination with CD28 costimulation (Figure 3), we additionally analyzed the transcriptional states of Tn cells activated for 24 h with a combination of αCD3+αCD28 and two different doses of CD70DT. Seurat clustering identified a total of 6 subclusters (Figure S6A). Subcluster 1 that was represented by resting Tn cells was connected via subcluster 2 with the remaining subclusters 3–6. While subcluster 6 was dominated by αCD3+αCD28 bead-activated cells, subcluster 3 was dominated by αCD3+CD70DT-activated cells. T cells activated with αCD3+αCD28 and high CD70DT dose, were mainly found in subclusters 4 and 5 which aligned in between subclusters 3 and 6. Tn cells activated with αCD3+αCD28 and low CD70DT dose were increasingly populated in the αCD3+αCD28 bead dominated cluster 6 (Figure S6B). Pseudotime-analysis revealed that resting Tn-associated signature genes such as FOXO1, FOXP1, TCF7, LEF1, IL7R and ZBTB20 showed a stepwise decrease of expression with transition from cluster 3 to 6. CD27 costimulation-associated signature genes (i.e. CCR7) were highest maintained in clusters 3 and 4, which was dominated by cells activated with αCD3+CD70DT or with high-dose CD70DT combined with αCD28. GAPDH, IL2RA and ENO1 genes associated with effector differentiation were increasingly induced from clusters 3 to 6 (Figure S6C). We then asked whether these early transcriptional changes observed in response to CD27 costimulation would be reflected in a spectrum of effector versus memory cell surface phenotypes after 9 days in culture. Increasing the dose of CD27 costimulation over αCD3+αCD28 stimulation promoted the maintenance of TCF1+CD27+ cells with a CD45RAhiCD45ROlowCD25lowCD95hi Tcm and Tscm phenotype (Figure S6D), but still activation with αCD3+CD70DT alone resulted in highest recovery of T cells with memory characteristics.
CD27 costimulation improves therapeutic efficacy of CAR-T cells
The differentiation state and metabolic fitness of T cells has been shown to affect their persistence and antitumor activity after adoptive transfer 14,15. We first used a murine model to examine whether CD27 signaling affected the ability of T cells to sustain function under conditions of chronic antigen stimulation. As described for human CD8+ T cells (see Figure 1), αCD3 and CD70DT stimulation of murine CD8+ T cells induced rapid CD27 surface receptor downmodulation (Figure S7A). We generated murine CD19-targeting 41BB_CD3ζ CAR-T cells using αCD3+CD70DT or αCD3+αCD28 stimulation, adoptively transferred these into immunocompetent mice that were monitored for B cell aplasia, CAR-T cell persistence and phenotype (Figure 6A). Despite reaching only 50% of the peak frequency of αCD3+αCD28-stimulated CAR-T cells after adoptive transfer, αCD3+CD70DT-stimulated CAR-T cells maintained B cell aplasia in more than 80% of mice, and persisted at high levels in blood compared to mice treated with αCD3+αCD28-stimulated CAR-T cells, which maintained B cell aplasia in only 40% of mice and persisted at lower levels at late time points (Figures 6B–6D). As early as 4 days after adoptive transfer, αCD3+CD70DT-stimulated CAR-T cells present in BM and spleen expressed higher levels of TCF1 and had a lower fraction of Ki67+ cells compared to those stimulated with αCD3+αCD28 (Figures 6E, 6F), and this difference in the Ki67+ fraction was maintained in the BM through day 21 (Figure 6G). Fewer αCD3+CD70DT-stimulated CAR-T cells in BM and spleen co-expressed inhibitory and activation markers (PD1+CD39+, PD1+TIM3+, PD1+TIGIT+) compared to αCD3+αCD28-stimulated T cells (Figure S7B). These data show that in this model of chronic antigen stimulation, αCD3+CD70DT-stimulated CD19 CAR-T cells more durably eliminate endogenous CD19+ B cells than αCD3+αCD28-stimulated CAR-T cells.
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) EGFRt−CD45.1+ 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.
We next evaluated the therapeutic activity of human CD19-targeting CAR-T cells in a human tumor xenograft model. 4-1BB_CD3ζ CD19 CAR-T cells were derived from unselected bulk CD8+ T cells after activation with plate-coated αCD3+CD70DT or with αCD3+αCD28 beads, the latter approach commonly used for stimulation in clinical CAR-T cell therapy. Cohorts of NSG mice were inoculated with CD19+ ffluc+ Raji lymphoma cell line and treated 7 days later with either αCD3+CD70DT CAR-T cells or αCD3+αCD28 CAR-T cells at a sub-curative dose (Figure 6H). In contrast to the murine chronic antigen stimulation model, αCD3+CD70DT-stimulated CAR-T cells expanded to higher numbers than αCD3+αCD28 CAR-T cells in this tumor model (Figure 6I). Before reaching the peak of expansion, αCD3+CD70DT-stimulated CAR-T cells expressed increased levels of PD-1 but lower levels of inhibitory markers TIM3 and TIGIT (Figure 6J) which resulted in more effective elimination of Raji tumor cells (Figure 6K).
The superior efficacy targeting hematopoietic cells in murine and human models suggested that CD27 signaling might generate CAR-T cells that would be more effective against solid tumors. To assess the ability of αCD3+CD70DT to sustain killing of solid tumor cells we stimulated ROR1-specific 4-1BB_CD3ζ CAR-T cells with αCD3+CD70DT or αCD3+αCD28 and performed serial restimulations with ROR1+ H1703 tumor cells without adjustment of E:T ratios (Figures S8A, S8B). αCD3+CD70DT CAR-T cells exhibited improved serial killing capacity compared to αCD3+CD28 CAR-T cells, despite showing reduced fold expansion, implicating an improved per-cell cytolytic capacity (Figures S8C, S8D). Analysis of CAR-T cells after the 4th re-stimulation revealed that αCD3+CD70DT CAR-T cells maintained higher cytokine secretion, expressed elevated levels of the cytolytic and activation markers GZMB and 4-1BB, and acquired a less exhausted phenotype (Figures S8E, S8F). We then evaluated ROR1 directed αCD3+CD70DT and αCD3+CD28 CAR-T cells in a ROR1+ breast-cancer xenograft model. Cohorts of NSG mice were inoculated with ROR1+ ffluc+ MDA-MB-231 cell line and treated 7 days later with Tn-derived CAR-T cells (Figure S8G). Mice receiving αCD3+CD70DT CAR-T cells showed prolonged tumor control whereas progressive tumor outgrowth was observed in αCD3+CD28 CAR-T cell treated mice around 18 days post infusion (Figure S8H). Moreover, CAR-T cell expansion was higher in mice treated with αCD3+CD70DT CAR-T cells compared to αCD3+CD28 CAR-T cells (Figure S8I). Taken together, these data show that αCD3+CD70DT-stimulated CAR-T cells possess a superior ability to sustain killing and function compared with αCD3+αCD28-stimulated CAR-T cells, as evaluated in the context of chronic and acute antigen exposure models.
Discussion
CD27 is expressed on T cells and engaged during an immune response by APCs that transiently express CD70 9. Signaling is further regulated by extracellular CD27 receptor cleavage that occurs beginning 24 h after ligand binding 17,18. The importance of regulating cellular responses mediated by CD27 is illustrated by the observation that transgenic mice in which CD70 is constitutively expressed in B cells exhibit T cell effector differentiation, exhaustion and lethal immunodeficiency 48. Our studies using a synthetic trimeric CD70 ligand to transiently bind CD27 during Tn cell activation in vitro demonstrate that CD27 is rapidly internalized by after ligand engagement by CME, identifying an immediate ligand-dependent mechanism to regulate the duration of CD27 engagement. Rapid internalization of CD27 was observed in co-cultures with CD70 expressing artificial APCs and with mature BM-derived DCs presenting antigen on MHC molecules, demonstrating the physiological relevance of rapid ligand induced CD27 endocytosis. CD27 expressed in more differentiated memory T cell subsets was also internalized, although the magnitude of endocytosis was reduced, suggesting that CD27 costimulation may be modulated differently in these subsets.
We found that ligating CD27 with higher doses of CD70DT led to more TRAF2 being bound to CD27, which may provide a scaffold to build the receptor signalosome, like that described for TNFR 4-1BB 49. Our studies show that TRAF2 is associated with SHP-1 and that this complex modulates Tn cell activation when CD28 is engaged by inhibiting phosphorylation of Lck and reducing phosphorylation of ERK1/2 and AKT. This finding has parallels to the observation using a cell-free membrane reconstitution system that activation of PD-1 and SHP-2 inhibits T cells by de-phosphorylation of CD28 and Lck 50. Our data shows that the CD27-TRAF2-SHP-1 axis modulates the strength of activation induced by TCR and CD28 signals only when CD70-mediated CD27 engagement is sufficient to induce rapid CD27 endocytosis, preserving the ability of primed Tn cells to differentiate along a memory rather than effector fate. Notably, when activated Tn cells received a dose of CD70DT that was insufficient to induce strong endocytosis, ERK1/2 phosphorylation and T cell proliferation was enhanced. This data suggests that alternative signaling mechanisms are activated depending on the level of CD27 engagement.
CD27 costimulation transiently delayed cell-cycle progression and altered metabolic and cell surface phenotype of T cells towards a Tscm and Tcm phenotype. This finding is in line with the observations that the formation of memory CD8+ T cells is marked by their slow replicative history 51,52. As early as 24 h after Tn cell activation, CD27 signaling promoted a distinct transcriptional and epigenetic profile characterized by reduced expression of genes associated with effector differentiation, contrasting that observed with CD28 costimulation. T cells receiving TCR, CD28 and CD27 signals adopted a transcriptional profile that was intermediate to that observed with αCD3+CD70DT or αCD3+αCD28 stimulation, implying that the strength of these signals cooperate to diversify T cell fates. 24 h CD27-costimulated T cells showed high utilization of transcription factors FOXO1, FOXP1, TCF1 and LEF1, which are amongst the top 5 most rapidly turned over TFs in quiescent Tn cells 53. This suggests that induction of high TCF7 transcript levels in response to αCD3+CD70DT activation may allow maintenance of high TCF1 protein levels despite high turnover of this TF. In contrast, αCD3+αCD28-activated Tn cells may fail to maintain TCF1 protein levels due to lower TCF7 transcript expression, despite having broadly accessible DNA binding motifs. FOXO1 is known to constrain CD8+ Tn cell activation by attenuating AP-1 effector programs 54 and is required for formation of Tcm in part by regulating the TCF7 gene locus 46. Further, our findings raise the question whether the observed CD27 signaling-induced FOXP1 regulatory network is conserved across T cell lineages, given that CD4+ Treg cell differentiation and identity are associated with both CD27 signaling 55 and FOXP1 transcriptional regulation 56.
Our data illustrating that CD27 costimulation of αCD3-activated Tn cells promotes the acquisition of memory properties led us to evaluate a new strategy for manufacturing of therapeutic T cell products. Disease relapse after CAR-T cell therapy targeting CD19 or BCMA can occur due to lack of CAR-T cell persistence and/or function, which in some cases may reflect a suboptimal quality of the expanded CAR-T infusion product 15. By engaging the CD27 costimulatory receptor, we were able to enrich the infusion product with T cells characterized by a less effector-differentiated state, which in turn persisted better under chronic antigen stimulation and mediated superior antitumor effects in vivo compared to αCD3+αCD28-activated CAR-T cells. Thus, CD27 costimulation could provide a qualitative advantage during CAR-T cell manufacturing and improve long-term CAR-T cell therapeutic efficacy.
Limitations of the study
Our data shows that CD28 is required for the modulatory effects of CD70DT on Lck and ERK1/2 phosphorylation in activated Tn cells but does not address whether the affected Lck is associated with CD28 or with other binding partners. It is also unknown whether varlilumab, an agonistic CD27 mAb that has been in the clinic, would have similar effects to the natural CD70 ligand. Endocytosis of CD27, which is greater with more engagement by CD70DT, is required for the effects on LCK and ERK1/2 phosphorylation, however we did not evaluate alternative signaling mechanisms from non-endocytosed CD27 when a lower amount of CD27 is engaged by CD70DT. Although we demonstrate that the level CD70 on murine DC and CD70-transgene expressing K32 cells is sufficient for CD27 internalization, a limitation of our study is that the signaling work was largely performed using CD70DT and agonistic antibodies. Additional studies examining the consequences of CD70 interactions with CD27 in vivo are necessary. We compared αCD3+αCD28 beads to αCD3+CD70DT for generating CAR-T cells because we observed the greatest preservation of memory properties with αCD3+CD70DT compared to activation conditions that included αCD28. Although efficacy of CAR-T cells has been associated with memory properties, the active ingredient(s) in heterogeneous T cell products that confer treatment efficacy after adoptive cell therapy are incompletely defined. Other stimulation conditions for generating therapeutic T cells that include different doses of CD70DT and αCD28 might be beneficial and should be evaluated in future studies. Finally, the Multi-ome analysis of Tn cells receiving αCD3+αCD28, αCD3+αCD28+CD70DT, and αCD3+CD70DT stimulation was performed at a single time point. This analysis captures early differences resulting from these different activation signals but does not describe differences in transcriptional or epigenetic profiles that may emerge later.
STAR Methods
RESSOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Stanley R. Riddell (sriddell@fredhutch.org).
Materials availability
Requests for in house synthesized proteins should be directed to the lead contact, Stanley R. Riddell (sriddell@fredhutch.org).
Data and code availability
Multiome-seq data have been deposited at the GEO repository and are publicly available as of the date of publication. Accession number is listed in the key resources table. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
Key resources table.
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Antibodies | ||
| THE™ HIS Tag Antibody – iFluor 647 (1:200) | GenScript | Cat#A01802; RRID: AB_2943644 |
| Brilliant Violet 785 anti-human CD45 (clone HI30, 1:100) | BioLegend | Cat#304031; RRID: AB_10900423 |
| BD Horizon BV421 anti-human CD45RO (clone UCHL1, 1:100) | BD Biosciences | Cat#562641; RRID: AB_2737696 |
| BD Horizon BUV395 anti-human CD45RO (clone UCHL1, 1:40) | BD Biosciences | Cat#564292; RRID: AB_2744410 |
| Alexa Fluor 700 anti-human CD45RA (clone HI100, 1:100) | BioLegend | Cat#304119; RRID: AB_493762 |
| PerCP/Cyanine5.5 anti-human CCR7 (clone G043H7, 1:100) | BioLegend | Cat#353219; RRID: AB_10915275 |
| Brilliant Violet 785 anti-mouse/rat/human CD27 (clone LG3.A10, 1:800) | BioLegend | Cat#124241; RRID: AB_2800595 |
| Brilliant Violet 510 anti-mouse/rat/human CD27 (clone LG3.A10, 1:200) | BioLegend | Cat#124229; RRID: AB_2565795 |
| APC/Cyanine7 anti-human CD27 (clone MT-271, 1:100) | BioLegend | Cat#356423; RRID: AB_2566772 |
| FITC anti-human CD27 (clone O323, 1:100) | BioLegend | Cat#302805; RRID: AB_314297 |
| PerCP/Cyanine5.5 anti-human CD70 (clone 113-16, 1:100) | BioLegend | Cat#355107; RRID: AB_2562478 |
| eFluor 450 anti-human CD69 Monoclonal Antibody (clone FN50, 1:100) | eBioscience | Cat#48-0699-42; RRID: AB_2574025 |
| PE/Cyanine7 anti-human CD137 (clone 4B4-1, 1:100) | BioLegend | Cat#309817; RRID: AB_2287731 |
| BD OptiBuild BUV737 mouse anti-human CD137 (clone 4B4-1, 1:100) | BD Biosciences | Cat#741861; RRID: AB_2871191 |
| APC anti-human CD25 (clone BC96, 1:200) | BioLegend | Cat#302609; RRID: AB_314279 |
| Pacific Blue anti-human CD127 (clone A019D5, 1:50) | BioLegend | Cat#351305; RRID: AB_10718638 |
| Brilliant Violet 605 anti-human CD95 (clone DX2, 1:100) | BioLegend | Cat#305627; RRID: AB_2562444 |
| FITC anti-human CD8α (clone RPA-T8, 1:500) | BioLegend | Cat#301005; RRID: AB_314123 |
| BD Horizon BUV496 mouse anti-human CD8 (clone RPA-T8, 1:500) | BD Biosciences | Cat#612943; RRID: AB_2916884 |
| PE anti-human CD279 (PD-1) Monoclonal Antibody (clone eBioJ105, 1:100) | eBioscience | Cat#12-2799-42; RRID: AB_11042478 |
| PE-Cyanine7 anti-human CD279 (PD-1) Monoclonal Antibody (clone eBioJ105, 1:100) | eBioscience | Cat#25-2799-42; RRID: AB_10853804 |
| BD Horizon PE-CF594 mouse anti-human TIM-3 (clone 7D3, 1:100) | BD Biosciences | Cat#565561; RRID: AB_2744371 |
| Alexa Fluor 647 anti-human TIGIT (clone A15153G, 1:100) | BioLegend | Cat#372723; RRID: AB_2715971 |
| Brilliant Violet 605 anti-human CD223 (LAG-3) (clone 11C3C65, 1:100) | BioLegend | Cat#369324; RRID: AB_2721541 |
| Brilliant Violet 711 anti-human CD39 (clone A1, 1:100) | BioLegend | Cat#328227; RRID: AB_2632893 |
| PE anti-human EGFR (clone AY13, 1:100) | BioLegend | Cat#352903; RRID: AB_10898161 |
| APC anti-human EGFR (clone AY13, 1:100) | BioLegend | Cat#352905; RRID: AB_11148943 |
| APC-Cyanine7 anti-human CD62L (clone DREG-56, 1:100) | BioLegend | Cat#304813; RRID: AB_493583 |
| FITC anti-human CD19 (clone HIB19, 1:100) | BioLegend | Cat#302205; RRID: AB_314235 |
| PE anti-human CD19 (clone HIB19, 1:100) | BioLegend | Cat#302207; RRID: AB_314237 |
| BD Horizon BV421 anti-human Granzyme B (clone GB11, 1:100) | BD Biosciences | Cat#563389; RRID: AB_2738175 |
| PE mouse anti-TCF-7/TCF-1 (clone S33-966, 1:100) | BD Biosciences | Cat#564217; RRID: AB_2687845 |
| PE mouse IgG1k Isotype Control | BD Biosciences | Cat#554680; RRID: AB_395506 |
| APC anti-human IL-2 (clone MQ1-17H12, 1:100) | eBioscience | Cat#17-7029-82; RRID: AB_469492 |
| PE-Cyanine7 anti-human TNF-α (clone Mab11, 1:100) | BioLegend | Cat#502929; RRID: AB_2204080 |
| PE anti-human IFN gamma (clone 4S.B3, 1:100) | eBioscience | Cat#12-7319-42; RRID: AB_1311247 |
| PE-Cy7 anti-mouse Ki-67 (clone B56, 1:100) | BD Biosciences | Cat#561283; RRID: AB_10716060 |
| PE-Cyanine7 anti-mouse CD39 (clone Duha59, 1:100) | BioLegend | Cat#143805; RRID: AB_2563393 |
| Alexa Fluor 700 anti-mouse CD45.1 (clone A20, 1:200) | BioLegend | Cat#110723; RRID: AB_493732 |
| Brilliant Violet 605 anti-mouse CD279 (PD-1) (clone 29F.1A12, 1:100) | BioLegend | Cat#135219; RRID: AB_11125371 |
| BV711 mouse anti-mouse TIGIT (clone 1G9, 1:100) | BD Biosciences | Cat#744214; RRID: AB_2742063 |
| Brilliant Violet 421 anti-mouse CD366 (TIM-3) (clone RMT3-23, 1:100) | BioLegend | Cat#119723; RRID: AB_2616908 |
| BUV661 anti-mouse CD19 (clone 1D3, 1:200) | BD Biosciences | Cat#612971; RRID: AB_2870243 |
| Brilliant Violet 785 anti-mouse CD223 (LAG-3) (clone C9B7W, 1:100) | BioLegend | Cat#125219; RRID: AB_2566571 |
| BUV395 rat anti-mouse CD8a (clone 53-6.7, 1:500) | BD Biosciences | Cat#565968; RRID: AB_2739421 |
| Pacific Blue rat anti-mouse CD8a (clone 53-6.7, 1:500) | BD Biosciences | Cat#558106; RRID: AB_397029 |
| BUV496 rat anti-mouse CD4 (clone GK1.5, 1:200) | BD Biosciences | Cat#612952; RRID: AB_2813886 |
| PE/Cyanine7 anti-mouse CD70 (clone FR70, 1:300) | BioLegend | Cat#104611; RRID: AB_2750466 |
| FITC anti-mouse CD11c (clone N418, 1:300) | BioLegend | Cat#117305; RRID: AB_313774 |
| BV421 rat anti-mouse I-A/I-E (clone M5/114.15.2, 1:400) | BD Biosciences | Cat#562564; RRID: AB_2716857 |
| APC anti-mouse CD80 (clone 16-10AL, 1:300) | BioLegend | Cat#104713; RRID: AB_313134 |
| Alexa Fluor 700 anti-mouse CD86 (clone GL-1, 1:200) | BioLegend | Cat#105023; RRID: AB_493720 |
| BV711 mouse Anti-Ki-67 (clone B56, 1:100) | BD Biosiences | Cat#563755; RRID: AB_2738406 |
| PE anti-human ERK1/2 phospho [T202,Y204] (clone 6B8B69, 1:30) | BioLegend | Cat#369506; RRID: AB_2629705 |
| Alexa Fluor 488 anti-human Akt phospho [Thr308] (clone D25E6, 1:50) | Cell Signaling Technology | Cat#43506; RRID: AB_2799244 |
| Alexa Fluor 488 anti-human SHP-1 Rabbit mAb (clone E1U6R, 1:2000) | Cell Signaling Technology | Cat#77181; RRID: AB_2943643 |
| Alexa Fluor 488 Rabbit mAb IgG XP Isotype Control (clone DA1E, 1:1000) | Cell Signaling Technology | Cat#2975; RRID: AB_10699151 |
| Pacific Blue mouse anti-human CD8 (clone RPA-T8, 1:100) | BD Biosciences | Cat#558207; RRID: AB_397058 |
| InVivoMAb anti-mouse CD70 | BioXCell | Cat#BE0022; RRID: AB_1107667 |
| InVivoMAb rat IgG2b isotype control | BioXCell | Cat#BE0090; RRID: AB_1107780 |
| TotalSeq™-A0251 anti-human Hashtag 1 Antibody | BioLegend | Cat#394601; RRID: AB_2750015 |
| TotalSeq™-A0252 anti-human Hashtag 2 Antibody | BioLegend | Cat#394603; RRID: AB_2750016 |
| TotalSeq™-A0253 anti-human Hashtag 3 Antibody | BioLegend | Cat#394605; RRID: AB_2750017 |
| TotalSeq™-A0254 anti-human Hashtag 4 Antibody | BioLegend | Cat#394607; RRID: AB_2750018 |
| TotalSeq™-A0255 anti-human Hashtag 5 Antibody | BioLegend | Cat#394609; RRID: AB_2750019 |
| TotalSeq™-A0256 anti-human Hashtag 6 Antibody | BioLegend | Cat#394611; RRID: AB_2750020 |
| TotalSeq™-A0257 anti-human Hashtag 7 Antibody | BioLegend | Cat#394613; RRID: AB_2750021 |
| TotalSeq™-A0258 anti-human Hashtag 8 Antibody | BioLegend | Cat#394615; RRID: AB_2750022 |
| Anti-human CD27 (clone E6S8H) Rabbit mAb | Cell Signaling Technology | Cat#28034; RRID: AB_2943642 |
| Anti-human β2-microgloblulin (clone D8P1H) Rabbit mAb | Cell Signaling Technology | Cat#12851; RRID: AB_2716551 |
| Anti-human β–Actin (clone 13E5) Rabbit mAb | Cell Signaling Technology | Cat#4770; RRID: AB_2223172 |
| Anti-human GAPDH (clone D16H11) XP Rabbit mAb | Cell Signaling Technology | Cat#5174; RRID: AB_10622025 |
| Anti-human TRAF2 (clone C192) Rabbit mAb | Cell Signaling Technology | Cat#4724; RRID: AB_2209845 |
| Anti-human SHP-1 (clone E1U6R) Rabbit mAb | Cell Signaling Technology | Cat#26516; RRID: AB_2934293 |
| Anti-human SHP-1 phospho [Tyr564] (clone D11G5) Rabbit mAb | Cell Signaling Technology | Cat#8849; RRID: AB_11141050 |
| Anti-human SH-PTP-2 (clone B-1) Mouse mAb | Santa Cruz Biotechnology | Cat#sc-7384; RRID: AB_628252 |
| Anti-human SHP1 phospho [Tyr536] | Abcam | Cat#ab41437; RRID: AB_777914 |
| Anti-human SHP1 phospho [Tyr591] | Abcam | Cat#ab41436; RRID: AB_777913 |
| Anti-human Lck (clone D88) XP Rabbit mAb | Cell Signaling Technology | Cat#2984; RRID: AB_2136313 |
| Anti-human phospho Lck [Tyr394] (clone E5L3D) Rabbit mAb | Cell Signaling Technology | Cat#70926; RRID: AB_2924371 |
| Anti-human phospho Lck [Tyr505] | Cell Signaling Technology | Cat#2751; RRID: AB_330446 |
| Anti-mouse IgG, HRP-linked Antibody | Cell Signaling Technology | Cat#7076; RRID: AB_330924 |
| Anti-rabbit IgG, HRP-linked Antibody | Cell Signaling Technology | Cat#7074; RRID: AB_2099233 |
| Bacterial and virus strains | ||
| Turbo Competent E. coli (MAX Efficiency DH5α) | Invitrogen | Cat#18258-012 |
| Biological samples | ||
| Human CD8+ T cells isolated from PBMCs | Fred Hutchinson Cancer Center / Bloodworks Northwest | N/A |
| Chemicals, peptides, and recombinant proteins | ||
| ES91-17, 100 μM | Sigma-Aldrich | Cat#SML2712; CAS: 55854-43-8 |
| Mdivi-1, 50 μM | Selleck | Cat#S7162; CAS: 338967-87-6 |
| Latrunculin A, 0.04 μM | Sigma-Aldrich | Cat#76343-93-6; CAS: 76343-93-6 |
| Dynasore, see titration | Selleck | Cat#S8047; CAS: 304448-55-3 |
| Dyngo-4a, see titration | Selleck | Cat#S7163; CAS: 1256493-34-1 |
| ULK-101, 5 μM | Selleck | Cat#S8793; CAS: 2443816-45-1 |
| Lactacystin, 20 μM | Tocris | Cat#2267; CAS: 133343-34-7 |
| TPI-1, 0.4 μM | Selleck | Cat#S6570; CAS: 79756-69-7 |
| GM6001, 10 μM | Selleck | Cat#S7157; CAS: 142880-36-2 |
| MitoTracker™ Green, 100 nM | ThermoFisher | Cat#M7514 |
| CellTrace™ CFSE dye, 0.1 μM | ThermoFisher | Cat#C34554 |
| 7-AAD | Sigma-Aldrich | Cat#SML1633 |
| Recombinant human IL-2 (clinical grade Proleukin®, Aldesleukin) | Prometheus Laboratories | N/A |
| Recombinant human IL-7 | PeproTech | Cat#200-07; GenPept: P13232 |
| Recombinant mouse GM-CSF | PeproTech | Cat#315-03; GenPept: P01587 |
| Ultra-LEAF™ Purified anti-human CD3 (clone OKT3) | BioLegend | Cat#317347 RRID: AB_2571994 |
| Anti-human CD28 (clone TGN1412) | This paper | N/A |
| Ultra-LEAF™ Purified anti-human CD28 (clone CD28.2) | BioLegend | Cat#302934 RRID: AB_11148949 |
| Anti-human CD70 Dimer Trimer (CD70DT) | This paper | N/A |
| IgG1-Fc | This paper | N/A |
| Purified NA/LE Hamster Anti-Mouse CD28 (clone 37.51) | BD Pharmingen | Cat#553294 RRID: AB_394763 |
| Purified NA/LE Hamster Anti-Mouse CD3ε (clone 145-2C11) | BD Pharmingen | Cat#553057 RRID: AB_394590 |
| Dynabeads™ Human T-Activator CD3/CD28 | Gibco | Cat#11131D |
| Critical commercial assays | ||
| Chromium Next GEM Single Cell Multiome ATAC + Gene Expression Reagent Bundle | 10X Genomics | Cat#1000258 |
| Chromium Next GEM Chip J Single Cell Kit | 10X Genomics | Cat#1000230 |
| Chromium Next GEM Chip G Single Cell Kit | 10X Genomics | Cat#1000127 |
| CalPhos Mammalian Transfection Kit | Clonetech / Takara | Cat#631312 |
| Seahorse XF Glycolysis Stress Test Kit | Agilent | Cat#103020-100 |
| Seahorse XF Mitochondrial Stress Test Kit | Agilent | Cat#103015-100 |
| P3 Primary Cell 4D-Nucleofector™ X Kit S | Lonza | Cat#V4XP-3032 |
| NucleoSpin RNA XS Micro kit for RNA purification | Macherey-Nagel | Cat#740902 |
| SuperScript IV First-Strand Synthesis System Kit | ThermoFisher | Cat#18091050 |
| EasySep™ Human Naïve CD8+ T Cell Isolation Kit II | STEMCELL | Cat#17968 |
| EasySep™ Human CD8+ T Cell Isolation Kit | STEMCELL | Cat#17953 |
| EasySep™ Mouse CD8+ T Cell Isolation Kit | STEMCELL | Cat#19853 |
| Pierce™ BCA Protein Assay Kit | ThermoFisher | Cat#23225 |
| Dynabeads™ Protein G Immunoprecipitation Kit | Invitrogen | Cat#10007D |
| Deposited data | ||
| Multiome Single Cell RNAseq data | This paper | GEO: GSE244834 |
| Experimental models: Cell lines | ||
| Lenti-X™ 293T cells | Clonetech / Takara | Cat#632180 RRID: CVCL_4401 |
| K32 | Carl June, Univerisy of Pennsylvania (Maus et al., 2002) | N/A |
| CD70-transgene expressing K32 | This paper | N/A |
| Raji | ATCC | Cat#CCL-86 RRID: CVCL_0511 |
| Raji+ffluc+GFP | This paper | N/A |
| MDA-MB-231+ffluc+GFP | This paper | N/A |
| NCI-H1703 | ATCC | Cat#CRL-5889 RRID: CVCL_1490 |
| NIH-3T3 | ATCC | Cat#CRL-1658 RRID: CVCL_0594 |
| CD40L-transgene expressing NIH-3T3 | This paper | N/A |
| Platinum-E cells | Cell Biolabs Inc. | Cat#RV-101 RRID: CVCL_B488 |
| Experimental models: Organisms/strains | ||
| Mouse: C57BL/6 wildtype (WT) | The Jackson Laboratory | RRID: IMSR_JAX:000664 |
| Mouse: B6.SJL-Ptprca Pepcb/BoyJ (CD45.1) | The Jackson Laboratory | RRID: IMSR_JAX:002014 |
| Mouse: C57BL/6-Tg(CAG-OVAL)916Jen/J (Act-mOVA) | The Jackson Laboratory | RRID: IMSR_JAX:005145 |
| Mouse: C57BL/6-Tg(TcraTcrb)1100Mjb/J (OT-1) | The Jackson Laboratory | RRID: IMSR_JAX:003831 |
| Mouse: NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) | The Jackson Laboratory | RRID: IMSR_JAX:005557 |
| Oligonucleotides | ||
| sgRNA targeting sequence: SHP-1 (PTPN6): 5’ [CAUCCAGCCCACUGAGGUCU] 3’ | Synthego | N/A |
| Primer: PTPN6_Ex2_Forward1: 5’ [CTCTCTGCCTGCCCAGACTA] 3’ | IDT | N/A |
| Primer: PTPN6_Ex2_Reverse1: 5’ [ACGGGGAACCAGGAATGAGTG] 3’ | IDT | N/A |
| TaqMan Gene Expression Assay: B2M (Hs00187842_m1) | ThermoFisher Scientific | Cat#4331182 |
| TaqMan Gene Expression Assay: CD27 (Hs00386811_m1) | ThermoFisher Scientific | Cat#4331182 |
| Recombinant DNA | ||
| Lentiviral Packaging Vector: pCHGP-2 | This paper | N/A |
| Lentiviral Packaging Vector: pCMV-Rev2 | This paper | N/A |
| Lentiviral Packaging Vector: pCMV-G | This paper | N/A |
| Plasmid: epHIV7_hCD19(FMC63)_IgG4_CD28TMD_h41BB_ hCD3ζ_T2A_EGFRt | This paper | N/A |
| Plasmid: epHIV7_hROR1(R12)_IgG4_CD28TMD_ h41BB_ hCD3ζ_T2A_CD19t | This paper | N/A |
| Plasmid: epHIV7_hROR1(R12)_IgG4_CD28TMD_ h41BB_ hCD3ζ_T2A_EGFRt | This paper | N/A |
| Plasmid: epMP71-mCD19(1D3)_IgG4D28TMD_m41BB_ mCD3ζ_T2A_mCD19t | This paper | N/A |
| Plasmid: epHIV7_hCD70 | This paper | N/A |
| Plasmid: ep-HIV7_mCD40L | This paper | N/A |
| Software and algorithms | ||
| FlowJo software v10.8.1 | Becton Dickinson & Company | https://www.flowjo.com/ |
| Thermo TraceFinder 4.1 | Thermo Fisher Scientific | https://www.thermofisher.com/us/en/home/industrial/mass-spectrometry/liquid-chromatography-mass-spectrometry-lc-ms/lc-ms-software/lc-ms-data-acquisition-software/tracefinder-software.html |
| FACSDIVA v9.1 | Becton Dickinson & Company | https://www.bdbiosciences.com/en-us/products/software/instrumentsoftware/bd-facsdiva-software |
| ID A v2.0 Analysis Software | SONY | N/A |
| Prism v9.4.0 | GraphPad | https://www.graphpad.com/features |
| Living Image Analysis Software | Perkin Elmer | http://www.perkinelmer.com/product/li-software-for-spectrum-1-seat-addon-128113 |
| R v4.2.0 | CRAN | https://cran.r-project.org/mirrors.html |
| RStudio v2022.02.3 | RStudio | https://rstudio.com/ |
| Cellranger ARC | 10X Genomics | https://support.10xgenomics.com/single-cell-multiome-atac-gex/software/downloads |
| Seurat | (Stuart et al., 2021) | https://github.com/satijalab/seurat |
| MACS2 | (Feng et al., 2012) | https://pypi.org/project/MACS2/ |
| Signac | (Stuart et al., 2021) | https://stuartlab.org/signac/ |
| Harmony | (Korsunsky et al., 2019) | https://portals.broadinstitute.org/harmony/index.html |
| ChromVAR | (Schep et al., 2017) | https://greenleaflab.github.io/chromVAR/index.html |
| FigR | (Kartha et al., 2022) | https://buenrostrolab.github.io/FigR/ |
| Monocle3 | (Trapnell et al., 2014) | https://cole-trapnell-lab.github.io/monocle3/ |
| Other | ||
| Trans-blot Turbo System | Bio-Rad | N/A |
| PVDF membranes | Bio-Rad | Cat#10026934 |
| Mini-PROTEAN® TGX™ Precast Protein Gels | Bio-Rad | Cat#456-1085 |
| ECL Chemiluminescent Substrate | Bio-Rad | Cat#1705062 |
EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
Mice
C57BL/6 wildtype (WT), B6.SJL-Ptprca Pepcb/BoyJ (CD45.1), C57BL/6-Tg(CAG-OVAL)916Jen/J (Act-mOVA), C57BL/6-Tg(TcraTcrb)1100Mjb/J (OT-1) and NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice were purchased from Jackson Laboratory and housed in specific-pathogen-free conditions. For all studies, 6–8 week old age-matched and sex-matched male or female mice were used. All mice were housed at the Fred Hutchinson Cancer Center (Seattle, WA). All experiments were performed with the approval of the Institutional Animal Care and Use Committee of the Fred Hutchinson Cancer Center and performed in accordance with institutional and national guidelines and regulations.
Human subjects
Peripheral blood mononuclear cells (PBMCs) were obtained from healthy human donors after written informed consent on research protocols approved by the Institutional Review Board (IRB) of the Fred Hutchinson Cancer Center (Seattle, WA). PBMCs were also purchased from Bloodworks Northwest.
Cell lines
293T Lenti-X cells (Clontech) were cultured in cDMEM (DMEM, 10% FBS, 1 mM L-glutamine, 25 mM Hepes, 100 U/mL penicillin+streptomycin). The K32 cell line was a kind gift from Carl H. June from the University of Pennsylvania 19. K32 cells were cultured in IMDM, 10% FBS and penicillin+streptomycin (100 U/mL; Gibco). K32 cells were lentiviral transduced with a human truncated CD70 transgene to generate CD70-transgene expressing K32 cells. CD19+ Raji lymphoma cells expressing firefly luciferase (Raji+ffluc), ROR1+ NCI-H1703 lung squamous carcinoma cells and ROR1+ MDA-MB-231 triple negative breast cancer cells expressing firefly luciferase (MDA-MB-231+ffluc) were purchased from ATCC and maintained in LCL medium (RPMI 1640, 10% FBS, 1 mM L-glutamine, 100 U/mL penicillin+streptomycin). NIH-3T3 fibroblasts were purchased from ATCC, maintained in cDMEM and lentiviral transduced with a murine CD154 (CD40L)-transgene and used to maturate murine BM derived DCs. Mycoplasma testing was performed bimonthly on all cell lines.
METHOD DETAILS
Recombinant protein design, production & modelling
CD70DT was constructed with human CD70 (Uniprot ID: P32970) amino acids Q39-P193. We introduced GS amino acids linking CD70 protomers to generate a single-chain trimer (scT). The designed CD70 scT was fused to human IgG1 Fc (Uniprot ID: P01857) amino acids E99-K330 with an introduced C103S mutation in the hinge region. The amino acid linker between the CD70 scT and IgG1-Fc was engineered to contain a TEV protease site, 6X Histidine purification tag, and an AviTag. The murine Ig kappa signal peptide was added to the N-terminus of the construct to facilitate secretion. For CD70DT and αCD28 (clone TGN1412) constructs, human codon sequence optimization and gene synthesis was carried out by Genscript. CD70DT and TGN1412 were expressed in suspension adapted FreeStyle 293-F cells (ThermoFisher, #R79007) using the Daedalus expression platform as described in 57 CD70DT and TGN1412 were purified from expression culture supernatant by HisTrap FF crude (Cytiva, #11000458) Ni-affinity chromatography or HiTrap MabSelect SuRe (Cytiva, 11003494) protein A affinity chromatography respectively. Following crude affinity purification, proteins were further purified by Superdex 200 (Cytiva, #28-9909-44) size exclusion chromatography. Purified proteins were flash frozen in liquid nitrogen in 1X PBS with 5% glycerol and stored at −80 °C.
The 3D model of CD70DT was generated with Colabfold 58 under default settings using the PDB70 reference database and including Amber side chain relaxation. CD70 scT with the linker region and Fc domains were modeled in Colabfold separately and then joined using Chimera X 59 to build the full-length model. Pymol (The PyMOL Molecular Graphics System, Version 2.0 Schrödinger, LLC) was used to generate the final images for publication.
CAR construction & virus production
Human CD19- and ROR1- specific CARs were constructed using VL and VH segments of the FMC63 60 and R12 61 mAbs respectively, linked by a human IgG4 spacer domain to a transmembrane domain of human CD28 (UniProt: P10747, aa153–179) and the cytoplasmic domains of human 4–1BB (UniProt: Q07011, aa 214–255) and human CD3ζ (UniProt: P20963, aa52–164). The construct encoded a ribosomal skip element (T2A) and a truncated human epidermal growth factor receptor (EGFRt) or CD19 receptor (CD19t). Transgenes were synthesized and cloned into epHIV7 lentiviral vectors. Lentivirus containing supernatants from 293T cells were produced using packaging vectors pCHGP-2, pCMV-Rev2 and pCMV-G, collected and concentrated prior to cryopreservation.
Mouse CD19-specific CARs were constructed using VL and VH segments of the 1D3 62 mAbs, linked by a mouse IgG4 spacer domain and a transmembrane domain of mouse CD28 (UniProt: P31041, aa151–177) to the cytoplasmic domain of mouse 4–1BB (UniProt: P20334, aa209–256) and mouse CD3ζ (UniProt: P24161, aa52–164). The construct encoded a ribosomal skip element (T2A) and a truncated human EGF Receptor (EGFRt). Transgenes were synthesized and cloned into pMP71 retroviral vectors and retrovirus containing supernatants were prepared from Platinum-E cells.
T cell isolation, activation & viral transduction
Human naïve or bulk CD8+ T cells were isolated from PBMC and leukopaks using the EasySep Human CD8+ T cell Isolation Kits (STEMCELL#17968 and STEMCELL#17953) and cryopreserved. Where indicated, CCR7+, naïve (Tn), central memory (Tcm) or effector memory (Tem) CD8+ cells were FACS-sorted prior to cryopreservation. For all metabolic readouts, freshly isolated or cultured cells were used. For all other experiments T cells were thawed and cultured in RPMI 1640 with 0.02% bovine serum albumin (BSA) for 2 h prior to activation. For activation, T cells were cultured in CTL media (RPMI 1640, 10% human serum, 1 mM L-glutamine, 100 U/mL penicillin+streptomycin, 50 μM β-mercaptoethanol), supplemented with 50 U/mL human IL-2 (Proleukin, Prometheus). Purified T cells were activated for 3 days using plate-bound αCD3 (OKT3, 1–5 μg/ml), αCD28 (TGN1412, 0.2–10 μg/ml), CD70DT (0.8–5 μg/ml) or Human T-Activator CD3+CD28 Dynabeads (BEAD, Thermo Fisher Scientific, 111.31D) at a 3:1 [BEAD:T cell] ratio. Lentiviral transduction of anti-human CD19 CAR (HIV7_FMC63_BBζ_EGFRt), anti-human ROR1 CAR (HIV7_R12_BBζ_CD19t) or anti-human ROR1 CAR (HIV7_R12_BBζ_EGFRt) encoding vectors was performed 24 h after T cell activation. CAR-transduced T cells were expanded for 10 days prior to adoptive transfer.
Mouse T cells were isolated from spleen and lymph nodes using the EasySep Mouse CD8+ T cell Isolation Kit (STEMCELL19853). Purified mouse T cells were activated for 2 days using plate-bound αCD3ε (145-2C11, 1 μg/ml) and αCD28 (37.51, 1 μg/ml) or CD70DT (1 μg/ml) and cultured in mTCM media (RPMI 1640, 10% FBS, 1 mM sodium pyruvate, 1 mM HEPES, 100 U/mL penicillin+streptomycin, 50 μM β-mercaptoethanol), supplemented with 50 U/mL IL2 (Peprotech #200-02) in the first 3 days and then switched to 50 U/mL IL15 for the following 2 days. Retroviral anti-murine CD19 CAR (MP71_1D3_BBζ_EGFRt) supernatant was spinoculated on retronectin-coated (Takara) plates, before addition and spin transduction of 24 h activated T cells. CAR-transduced T cells were expanded for 5 days prior to adoptive transfer.
Xenograft & B-cell aplasia models
Raji+ffluc lympoma cells (5×105) were inoculated by intravenous injection into age-matched (6–8 weeks old) NOD/SCID/γc−/− (NSG) male mice. MDA-MB-231+ffluc TNBC cells (1×106) were inoculated by sub-cutaneous injection into age-matched (6–8 weeks old) NSG male mice. Tumor growth was monitored at weekly intervals using in vivo bioluminescence imaging after luciferin administration using the Xenogen IVIS Imaging System (Caliper Life Sciences) and data analyzed using Live Image Software 4.7.2 (Caliper Life Sciences). Seven days after tumor inoculation, CD8+ anti-human CD19 or ROR1 CAR-T cells were injected retro-orbitally (r.o.) at a sub-curative dose of 0.7 × 106 cells per mouse or 8 × 106 cells per mouse. Peripheral blood was drawn weekly to monitor CAR-T cell expansion and further characterization.
B cell aplasia was induced in wildtype C57/BL6 mice by retro-orbital injection of 1 × 106 anti-murine CD19 CAR-T cells derived from splenic CD8+ T cells of CD45.1 congenic wildtype mice. T cells were administered 4 h after lymphodepletion using 200 mg/kg cyclophosphamide (Cy) administered intraperitoneally (i.p.) 63. B cell aplasia and CAR-T expansion and persistence was monitored weekly in the peripheral blood. Mice were harvested 4 days and 21 days post CAR-T cell infusion for analysis of CAR-T cells in BM and spleen.
Serial restimulation assay
The xCELLigence RTCA eSight (Agilent) platform was used to assess real time impedance-based killing capacity of CAR-T cells. ROR1+ H1703 tumor cells were seeded at a density of 3×104 cells per well in CTL low-dose hIL-2 (5 U/mL) and cultured for up to 24 h until cell index of 1–2 was reached. Transduction marker (CD19t) positive anti-human ROR1 (MP71_R12_BBζ_CD19t) CAR-T cells that were expanded for 10 days, purified via positive MACS selection using human CD19 MicroBeads (Miltenyi Biotec, #130-050-301) and added to the target cells at a 1:2 (E:T) ratio in CTL supplemented with hIL-2 (5 U/mL). Every other day, CAR-T cells were harvested, MACS-purified and re-plated without E:T ratio adjustment on a newly prepared plate containing ROR1+ H1703 tumor cells. After the 4th re-stimulation round, CAR-T cells were washed and used to assess surface marker expression and cytokine production capacity in response to PMA and ionomycin re-stimulation.
Flow cytometry
Cells were stained in Flow Buffer (PBS, 0.5% FBS, 3.5 μM EDTA) with antibodies to cell-surface markers for 30 min at 4 °C. For intracellular staining, cells were permeabilized for 30 min at room temperature in freshly prepared Perm Buffer (Foxp3 Transcription Factor Fixation kit, eBioscience, #00-5523-00) and incubated for up to 2 h at 4 °C with antibodies specific for cytokines or transcription factors, respectively. To stain for CD70DT binding, THE™-HIS-tag antibody was used. Fixable Viability Dyes were added prior to surface stain in PBS (ThermoFisher; Violet & Aqua; 1:500). Human and mouse reactive antibodies were purchased as specified in Key Ressourse Table. Corresponding isotype controls were added where indicated.
For the phospho protein flow, T cells were activated in antibody-coated flat-bottom-96-well plates in the presence of DMSO or SHP-1 inhibitor (TPI-1, Selleck, #S6570, 0.4 μM) or actin polymerization inhibitor (LatA, Sigma Aldrich, 0.05 μM). At respective timepoints, cells were immediately fixed using pre-warmed BD™ Cytofix Buffer (BD Pharmingen, #554655) and incubated for 10 min at 37 °C. Cells were then permeabilized in BD™ Phosflow Perm Buffer III (BD Pharmingen, #558050) for 30 min on ice. Subsequently cells were washed 3 times with Stain Buffer (BD Pharmingen, #554656) and incubated with surface marker, intracellular marker and phospho antibody cocktails for 30 min at RT.
Sample acquisition was performed on a BD Biosciences LSRII Fortessa or Symphony A5 instrument using the FACSDiva software (v9.1; BD Biosciences) or the SONY ID7000™ Spectral Cell Analyzer using the ID A analysis software (v2.0; SONY). Data were analyzed using FlowJo software (v10.8.1; BD Biosciences).
Generation of BM-derived DCs
Murine BM derived DCs were generated as previously described with adaptatioin.64,65 In brief, BM cells of Act-mOVA mice were harvested and cultured at a density of 2×106 cells per petri dish in R10 mDC medium (RPMI 1640, 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin+streptomycin, 50 μM β-mercaptoethanol), supplemented with 25 ng/mL murine GM-CSF (PeproTech). Media change was performed every other day over 10 days. For maturation, DCs were incubated overnight on CD40L-transduced NIH-3T3 fibroblasts at a ratio of 2:1 [DC:3T3]. The morning after, DCs were gently collected from the 3T3 monolayer and characterized by flow cytometry.
Immune co-culture with APCs
K32 or CD70-transgene expressing K32 cells were coated with αCD3 (OKT3, 5 μg/mL) ± αCD28 (TGN1412, 1 μg/mL) mAb for 15 min at 37 °C in RPMI. Following a wash step, αCD3 or αCD3+αCD28-coated K32 cells were co-cultured with human Tn cells at a 1:1 or 5:1 [aAPC:T cell] ratio in a V-bottom 96-well plate in the presence or absence of CME inhibitors (ES9–17 + Mdivi-1). At respective timepoints, cells were subsequently stained following the standard flow or phospho-flow protocols.
Alternatively, CD8+ T cells were isolated from spleens and LNs from OT-1 mice as described above. T cells were then co-cultured with BM derived Act-mOVA DCs at a [1:1] ratio in the presence of blocking FR70 or IgG antibody (20 μg/mL). At indicated timepoints, T cells were washed, Fc-block was added, and surface staining was performed.
Internalization & cleavage assay
Prior to activation, T cells were pre-incubated in RPMI 0.02% for 10 min at 37 °C with Dynasore (Selleck, #S8047, titration), Dyngo-4a (Selleck, #S7163, titration), ES9–17 (Sigma-Aldrich, #SML2712, 100 μM) and/or M-divi1 (Selleck, #S7162, 50 μM) to inhibit CME. Latrunculin A (Sigma Aldrich, #L5163, 0.04 μM) was added at the time of activation. GM6001 (Selleck, #S7157, 10 μM) was used to inhibit CD27 cleavage. T cells were seeded at a density of 1×105 cells per 96-well to the antibody-coated flat-bottom plates and centrifuged for 1 min at 400 × g. At indicated timepoints of activation, cells were removed from the activation plate, transferred to a new 96-U-bottom-well plate on ice and subsequently stained for surface marker expression at 4 °C for 30 min before analysis by flow cytometry.
Protein degradation assay
Prior to activation, T cells were pre-incubated with inhibitors in RPMI 0.02% for 10 min at 37 °C. The following inhibitors were used: ES9–17 (Sigma-Aldrich, #SML2712, 100 μM), Mdivi-1 (Selleck, # S7162, 50 μM), ULK-101 (Selleck, #S8793, 5 μM), Lactacystin (Tocris, #2267, 20 μM). T cells were seeded at a density of 1×105 cells per 96-well to the antibody-coated flat-bottom plates and immediately centrifuged for 1 min at 400 × g. After 10 min of activation, pre-warmed Cytofix Buffer (BD, #554655) was added to wells and incubated for additional 10 min at 37 °C. Cells were then transferred to a 96-U-bottom-well plate and permeabilized with freshly prepared Cytofix+Cytoperm solution of the FOXP3 staining kit (eBioscience, #00-5523-00) for 30 min at room temperature. Cells were washed in freshly prepared 1X PermBuffer (eBioscience, #00-5523-00) and stained for 2 h in 1X PermBuffer containing antibodies. Total cellular protein expression was assessed by flow cytometry.
Metabolic assays
To evaluate mitochondrial mass, cultured cells were washed in pre-warmed PBS and stained with Mitotracker Green (ThermoFisher, Cat#M7514, 100 nM) and surface marker antibodies for 15 min at 37 °C. Cells were washed with pre-warmed CTL medium and subsequently analyzed by flow cytometry. Seahorse assays were performed as described.66 Rehydrated Seahorse XF cartridges were incubated in XF calibrant for 1 h at 37 °C (no CO2) prior to starting the experiment. Human T cells (5 or 10 days post activation) were counted and the concentration was adjusted to 5×106 cell per mL in complete RPMI XF assay media (Agilent, Cat#103015-100). This media was supplemented with additional substrates for the Mito Stress Test (2 mM L-Glutamine, 1 mM Pyruvate, 10 mM Glucose, 50 U/mL hIL2) and for the Glucose Stress Test (2 mM L-Glutamine, 50 U/mL hIL2). T cells were added to plates coated with poly-L-lysine (Sigma Aldrich, Cat#P8920,100 μg/mL) at a density of 2×105 cells per well and centrifuged for 5 min at 400 × g to adhere the cells to the plate. Metabolic profiles were assessed by injection of oligomycin (1.5 μM), carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP, 0.5 μM), rotenone (0.5 μM), antimycin A (0.5 μM), glucose (10 mM), 2-deoxyglucose (2-DG, 50 mM). Data were generated using XF report generator software (Agilent).
Proliferation & cell-cycle analysis
Tn cells were resuspended in pre-warmed PBS containing 0.1 μM CellTrace™ CFSE dye (ThermoFisher) and labelled for 10 min in a 37 °C waterbath and vortexed at half-time. Labelling was quenched with FBS and T cells were washed 3 more times with pre-warmed CTL medium. Tn cells were activated in antibody-coated or BEAD containing flat-bottom-96-well plates (non-tissue culture treated) at a density of 105 cells per well in CTL + 50 U/mL hIL-2. CFSE dilution was assessed by flow cytometry 72 h post Tn cell activation.
Cell-cycle analysis was performed 48 h and 96 h post Tn cell activation. Cultured cells were washed in PBS and stained with surface antibodies in FACS buffer for 30 min at 4 °C. Cells were then permeabilized in ice-cold 70% ethanol and kept on ice for 1 h. After two consecutive wash steps, cells were stained in PFT buffer (PBS, 1% FBS, 0.25% Triton X-100) containing 25 mg/mL 7-AAD (Sigma-Aldrich) for 30 min at room temp in the dark. Cells were washed twice in FACS buffer and analyzed by flow cytometry.
Glucose tracing
Tn cells were harvested from donors and plated at 4×105 cells per 100 μl well of a pre-coated 96-well dish. Cells were grown in glucose free RPMI (Gibco, #11979020) supplemented with 10% dialyzed human serum, 1mM L-glutamine, 100 U/mL penicillin+streptomycin, 50 μM β-mercaptoethanol and 100 μM of [1,2-13C] Glucose (Cambridge Isotopes, CLM-504-PK). The supernatant was collected at the indicated time points, centrifuged, and frozen until extraction. Metabolites were extracted from 20 μl of each sample with 300 μL of 100% HPLC grade ethanol, centrifuged (17,000 × g, 10 min, 4 °C), moved to new Eppendorf tubes, lyophilized, and stored at 20 °C. Lyophilized samples were then resuspended in 80% HPLC grade methanol and HPLC grade water.
LC-MS metabolite quantification
Metabolite extracts were analyzed on a QExactive HF-X hybrid-Orbitrap mass spectrometer equipped with an Ion Max API source and HESI-II probe, coupled to a Vanquish Flex Binary UHPLC system (Thermo Scientific, Waltham, MA). A 1 μL sample injection was gradient eluted from a Millipore 2.1×150mm (5 μm particle size) ZIC-pHILIC column (Millipore, Burlington, MA) using a mobile phase gradient as follows: 0–20min: 85–20% “B”, followed by a hold at 20% “B” for 4 minutes, followed by a return to 85% “B” over 0.5 minutes, and finishing with a column re-equilibration at 85% “B” for 10 column volumes. The flow rate was at 150 μL/min, the autosampler compartment temperature was set to 10 °C, and the column temperature used was 30 °C. Mobile phase “A” consisted of 20mM ammonium carbonate and 0.1% ammonium hydroxide (v:v), while mobile phase “B” consisted of 100% acetonitrile. Metabolites were detected over a range of 70–1050 m/z in the MS1 full-scan mode in both positive and negative polarities. Data were collected in the profile mode at 240K resolution (@ 200 m/z) using an AGC target value of 3,000,000 and a maximum injection time of 250 mseconds, with a chromatographic peak width (FWHM) set at 10 seconds. The source parameters used with the HESI probe were as follows: sheath and aux gas flow rates at 40 and 20 units respectively, spray voltage of 2.5kV, a capillary temperature of 335 °C, and the aux. gas heater temperature was 350 °C. The ion funnel level was set at 40%.
Gene deletion in resting human CD8+ Tn cells
Naïve CD8+ Tn cells were freshly isolated from PBMCs as described above. To form the CRISPR-Cas9-sgRNA RNP complex, synthetic SHP-1 sgRNAs (Synthego) were incubated with Cas9 nuclease protein NLS (Horizon Discovery, Cat# CAS12206) at a ratio of [4:1] (160pmol sgRNA+ 40pmol Cas9) for 15 min at 37 °C and then kept at 4 °C until further processed. Per single reaction, 2×106 freshly isolated Tn cells were washed twice with pre-warmed PBS and resuspended in 20 μl of P3 buffer (Lonza, V4XP-3032). The RNPs where then mixed with the cell 20 μl of cell suspension and transferred into a 16-well reaction cuvette of the 4D-Nucleofector System (Lonza). Cells were nucleofected using program EH-100 on the 4D-Nucleofector system. Immediately after electroporation, 80 μl of pre-warmed complete T cell media (CTL) containing 0.5 ng/mL human IL-7 (Peprotech, 200–07) was added to each well in the cuvette and incubated for 15 min at 37 °C for recovery. A total of 100 μL cell suspension was then gently transferred into individual 48-wells and supplemented with 800 μL of CTL containing human IL-7. Half-media change was performed every other day which allowed resting Tn cells to maintained over a culture period of 8 days. SHP-1 knock-out efficiencies were assessed using intracellular staining for SHP-1 following the ERK1/2 phos-flow staining protocol and sanger sequencing (https://ice.synthego.com/#/analyze/results/nyhutj3v19gvwksh). For functional experiments SHP-1 sgRNA #8 was selected. Sequence of sgRNA #8 as well as sequencing primer pairs are provided in the key resources table.
Nuclei isolation & single-cell RNA + ATAC sequencing
Cryopreserved CD8+ Tn from n = 1 donor (previously processed as described above) were thawed and activated for 24 h with either αCD3+αCD28 Beads at a 3:1 [BEAD:T cell] ratio or with combinations of plate-bound αCD3 (5 μg/mL), αCD28 (0.2 μg/mL) and CD70DT (0.8 or 5 μg/mL). Additionally, CD8+ Tn (naïve) cells from the same donor were thawed and directly readied for nuclei isolation without activation. To minimize batch effects, cells from each stimulation condition were captured in a single lane using10x Genomics multiome chemistry. Prior to capture, cells were hashed using TotalSeq™-A HTO anti-ubiquitous cell-surface-epitope DNA-barcoded antibodies (BioLegend, see Key Resources Table), according to manufacturer’s protocol. The “hash-tagged” cells underwent a gentle fixation by incubation at room temperature in 1X PBS 0.1% formaldehyde for 10 min, then quenching with glycine at 0.125 M and immediately washing with ice cold 1X PBS (supernatant removed by spinning cells at 500 × g for 5 min). Next, nuclei preparation was performed according to manufacturer’s specification (document # CG000356 Rev B), with the following optimization step. The optimal permeabilization and lysis time was determined by incubating the samples into 100 μL chilled lysis buffer for 2, 3, 4, 5, or 6 min on ice, then proceeding to the washing step. The quality of obtained nuclei was assessed using Acridine Orange and Propidium Iodide staining (Nexcelom Viastain AOPI # CS2-0106-25 mL) as well as visualizing cell morphology under a microscope. The “2 min” mark was determined as the optimal permeabilization time with cell viability of 89–97% and cell integrity maintained as viewed under the microscope. After cell counting, equal numbers of nuclei from the samples were pooled to a single tube in 1X Diluted Nuclei Buffer (10x Genomics, PN-2000207) at ~3,500 nuclei per μL. The nuclei were then prepared for transposition according to the 10x Genomics Next GEM Single Cell Multiome ATAC + Gene Expression User Guide (document # CG000338 Rev D). Ten thousand nuclei were targeted for capture. Libraries were sequenced using an Illumina NextSeq 2000. 7862 and 11063 cells were included post quality control for downstream final analysis of the respective experiment.
Single-cell RNA & ATAC computational analysis
Illumina BCL files were demultiplexed and processed using cellranger ARC. ATAC peaks were called on the resulting fragments file using MACS2 67 with the parameters ‘--nomodel --extsize 200 --shift -100’. RNA counts and fragment reads in MACS2 peaks matrices were counted using Signac 68. Cells meeting the following thresholds were retained for downstream analysis: percent mitochondrial RNA reads < 15%; 3 < log10(ATAC counts) < 5; 3 < log10(RNA/UMI counts) < 4.5; Fraction of reads in peaks > 40%; nucleosome_signal < 1.5; TSS.enrichment > 1. RNA counts were normalized using SCT transform 69 prior to PCA dimensionality reduction, after which batch correction was performed using Harmony 70. ATAC counts underwent TFIDF/SVD 68 dimensionality reduction. An integrated UMAP embedding was created using the FindMultiModalNeighbors 71 function. Cell subtypes were classified using the standard Seurat workflow. Cis-regulatory elements were identified with the ‘LinkPeaks’ function and motif deviation scores were computed using ChromVAR 72. Target genes of highly utilized transcription factors were inferred and calculated using FigR 73. Pseudotime trajectory analysis was performed using the PlotGenesInPseudotime function in Monocle3 74.
Western blot assay
5×105 plate-activated CD8+ Tn cells were transferred to an Eppendorf tube and washed with ice-cold PBS. Cell pellets were then resuspended in ice-cold NP-40 lysis buffer (Invitrogen, #FNN0021) supplemented with protease inhibitor and phosphatase inhibitors (Thermo Scientific, #78430 and #78428). Lysates were vigorously vortexed and kept on ice for additional 10 min to allow complete lysis. Lysates were centrifuged for 15 min at 16000 × g and lysates subsequently transferred to new Eppendorf tubes. Protein concentrations were quantified using Pierce™ BCA Protein Assay Kit (Thermo Scientific, #23225). 4X Laemmli Sample buffer (Bio-Rad, #1610747) and NuPAGE Sample Reducing Agent (Invitrogen, # NP0009) were added to the protein lysate prior to denaturation at 95 °C for 5 min.
Gel electrophoresis was performed using Mini-PROTEAN® TGX™ Precast Protein Gels (Bio-Rad, #456-1085) in freshly prepared Tris+Glycine+SDS Running Buffer (Bio-Rad, #1610732). Transfer to PVDF membranes (Bio-Rad, #10026934) was performed in transfer buffer (Bio-Rad, #10026938) using Trans-blot Turbo System (Bio-Rad). For membrane blocking and antibody incubation blocking buffer (Bio-Rad, #12010020) was used. Membranes were incubated over night at 4 °C with primary antibodies listed in the Key Resources Table. Membranes were incubated with secondary HRP-linked antibodies for 1 h at room temperature. Membranes were incubated with ECL Chemiluminescent Substrate (Bio-Rad, #1705062) and developed using the iBright FL1000 gel imaging system (Invitrogen).
Immunoprecipitation assay
3–5×106 plate-activated CD8+ Tn cells were transferred to Eppendorf tube and washed with ice-cold PBS. Sample preparation was performed as described for Western Blot analysis. Lysates were kept on ice while Dynabeads Protein G (ThermoFisher, #10003D) were conjugated with antibody. 50 μl Dynabeads per reaction were transferred to an Eppendorf tube, washed with PBS and magnetized 3 times for 5 min. TRAF2 antibody (Cell Signaling, #4724) and SHP-1 antibody (#26516), were incubated with the Dynabeads for 1 h at RT under rotation. Dynabeads were then magnetized for 5 min and washed with PBS-Tween (0.02%) for 3 min 5 times. Dynabeads were then resuspended in freshly prepared 5 mM BS3 (ThermoFisher, #A39266) cross-linker solution and incubated for 30min under rotation. Subsequently, cross-linking reaction was quenched adding 1 M Tris-buffer to the Dynabead cross-linking solution at a final concentration of 35 mM. Dynabeads were magnetized for 5 min and washed 3 more times for 5 min. Cell lysates were now added to antibody-conjugated Dynabeads and kept under rotation overnight at 4 °C. After overnight incubation, Dynabeads were magnetized and un-bound supernatant fractions discarded. Antibody-captured proteins were eluted from Dynabeads in elution buffer consistent of Laemmli Sample Buffer (Bio-Rad, #1610747) and NuPAGE Sample Reducing Agent (Invitrogen, # NP0009) and boiled at 95 °C for 5 min. Gel electrophoresis and transfer were performed as described for Western Blot analysis.
Quantitative PCR
RNA was extracted using the NucleoSpin RNA XS Micro kit for RNA purification (Macherey-Nagel, # 740902.50) and cDNA synthesized using the SuperScript IV First-Strand Synthesis System kit (ThermoFisher, #18091050). Amplifications were performed for 45 cycles on a Quant Studio 5 Real-Time PCR system in a 10 μl reaction consisting of 2X TaqMan™ Universal Master Mix with UNG (Applied Biosystems, #4440046), 2–5ng of cDNA, and 20X TaqMan™ Gene Expression Assay probes (Applied Biosystems): B2M (Hs00187842_m1), CD27 (Hs00386811_m1). The cycle threshold (Ct) was determined using QuantStudio Analysis Software, normalized to housekeeping gene B2M, and the level of gene expression calculated using the comparative Ct method (2(Ĉt)). Log-2 fold-change (FC) was calculated between activated and non-activated (n.a., naive) T cell groups.
QUANTIFICATION AND STATISTICAL ANALYSIS
Statistical analysis
All data are represented as the mean values ± SEM. Statistical significance was determined by paired and unpaired two-tailed Student’s t-test, one-way ANOVA, tow-way ANOVA, Log-Rank (Mantel-Cox) test and Turkeys multiple comparison test as indicated in the figure legends using Prism software (GraphPad). Statistical significance was established at the levels of * p<0.05; ** p<0.005; *** p<0.0005; **** p<0.0001.
Supplementary Material
Highlights:
CD27 is rapidly endocytosed in activated T cells after binding its ligand CD70
CD27-TRAF2-SHP-1 modulates Lck phosphorylation in CD28 costimulated T cells
CD27 signaling promotes phenotypic and transcriptional adaptations of T cell memory
CAR-T cells generated with CD27 costimulation have superior antitumor efficacy
Acknowledgements
This work was supported by grants from the NIH CA18029 and CA114536 (SRR), NCI 5 P30 CA015704 Cancer Center Support Grant, and Lyell Immunopharma. We wish to thank Margot Pont, Sophia Schreiber and Naina Singhi for supportive discussions. Further we wish to thank Don Parilla, Ekram A. Gad and Latrice King from the Comparative Medicine Shared Resource team for the support with mouse experiments. This research was supported by the Cellular Imaging Shared Resource RRID:SCR_022609 of the Fred Hutch, University of Washington Cancer Consortium (P30CA015704). For technical support, we wish to thank Lena Schroeder, Hoku West-Foyle and Jin Meng from the Cellular Imaging Shared Resource team as well as Michele Black, Andrew Berger, Rebecca Reeves and Ben Janoschek from the Flow Cytometry Shared Resource team. Illustrated figures were created with BioRender.
Footnotes
Declaration of Interests
C.A.J, 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; is a founder of and holds equity in Lyell Immunopharma; and has served on the advisory boards for Adaptive Biotechnologies and Nohla. No potential conflicts of interest were disclosed by the other authors.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
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] [Google Scholar]
- 2.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] [Google Scholar]
- 3.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] [Google Scholar]
- 4.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 free article] [PubMed] [Google Scholar]
- 5.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 free article] [PubMed] [Google Scholar]
- 6.Hendriks J, Gravestein LA, Tesselaar K, van Lier RA, Schumacher TN, and Borst J (2000). CD27 is required for generation and long-term maintenance of T cell immunity. Nat Immunol 1, 433–440. 10.1038/80877. [DOI] [PubMed] [Google Scholar]
- 7.Liu W, Maben Z, Wang C, Lindquist KC, Li M, Rayannavar V, Lopez Armenta I, Nager A, Pascua E, Dominik PK, et al. (2021). Structural delineation and phase-dependent activation of the costimulatory CD27:CD70 complex. J Biol Chem 297, 101102. 10.1016/j.jbc.2021.101102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hintzen RQ, Lens SM, Lammers K, Kuiper H, Beckmann MP, and van Lier RA (1995). Engagement of CD27 with its ligand CD70 provides a second signal for T cell activation. J Immunol 154, 2612–2623. [PubMed] [Google Scholar]
- 9.Keller AM, Groothuis TA, Veraar EA, Marsman M, Maillette de Buy Wenniger L., Janssen H, Neefjes J., and Borst J. (2007). Costimulatory ligand CD70 is delivered to the immunological synapse by shared intracellular trafficking with MHC class II molecules. Proc Natl Acad Sci U S A 104, 5989–5994. 10.1073/pnas.0700946104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Akiba H, Nakano H, Nishinaka S, Shindo M, Kobata T, Atsuta M, Morimoto C, Ware CF, Malinin NL, Wallach D, et al. (1998). CD27, a member of the tumor necrosis factor receptor superfamily, activates NF-kappaB and stress-activated protein kinase/c-Jun N-terminal kinase via TRAF2, TRAF5, and NF-kappaB-inducing kinase. J Biol Chem 273, 13353–13358. 10.1074/jbc.273.21.13353. [DOI] [PubMed] [Google Scholar]
- 11.Jaeger-Ruckstuhl CA, Hinterbrandner M, Hopner S, Correnti CE, Luthi U, Friedli O, Freigang S, Al Sayed MF, Buhrer ED, Amrein MA, et al. (2020). TNIK signaling imprints CD8(+) T cell memory formation early after priming. Nat Commun 11, 1632. 10.1038/s41467-020-15413-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gattinoni L, Klebanoff CA, and Restifo NP (2012). Paths to stemness: building the ultimate antitumour T cell. Nat Rev Cancer 12, 671–684. 10.1038/nrc3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.June CH, O’Connor RS, Kawalekar OU, Ghassemi S, and Milone MC (2018). CAR T cell immunotherapy for human cancer. Science 359, 1361–1365. 10.1126/science.aar6711. [DOI] [PubMed] [Google Scholar]
- 14.Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O’Connor RS, Hwang WT, et al. (2018). Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 24, 563–571. 10.1038/s41591-018-0010-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Deng Q, Han G, Puebla-Osorio N, Ma MCJ, Strati P, Chasen B, Dai E, Dang M, Jain N, Yang H, et al. (2020). Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat Med 26, 1878–1887. 10.1038/s41591-020-1061-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Correnti CE, Hallinan JP, Doyle LA, Ruff RO, Jaeger-Ruckstuhl CA, Xu Y, Shen BW, Qu A, Polkinghorn C, Friend DJ, et al. (2020). Engineering and functionalization of large circular tandem repeat protein nanoparticles. Nat Struct Mol Biol 27, 342–350. 10.1038/s41594-020-0397-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hintzen RQ, de Jong R, Hack CE, Chamuleau M, de Vries EF, ten Berge IJ, Borst J, and van Lier RA (1991). A soluble form of the human T cell differentiation antigen CD27 is released after triggering of the TCR/CD3 complex. J Immunol 147, 29–35. [PubMed] [Google Scholar]
- 18.Hintzen RQ, Lens SM, Beckmann MP, Goodwin RG, Lynch D, and van Lier RA (1994). Characterization of the human CD27 ligand, a novel member of the TNF gene family. J Immunol 152, 1762–1773. [PubMed] [Google Scholar]
- 19.Maus MV, Thomas AK, Leonard DG, Allman D, Addya K, Schlienger K, Riley JL, and June CH (2002). Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4–1BB. Nat Biotechnol 20, 143–148. 10.1038/nbt0202-143. [DOI] [PubMed] [Google Scholar]
- 20.Schneider-Brachert W, Tchikov V, Neumeyer J, Jakob M, Winoto-Morbach S, Held-Feindt J, Heinrich M, Merkel O, Ehrenschwender M, Adam D, et al. (2004). Compartmentalization of TNF receptor 1 signaling: internalized TNF receptosomes as death signaling vesicles. Immunity 21, 415–428. 10.1016/j.immuni.2004.08.017. [DOI] [PubMed] [Google Scholar]
- 21.Schutze S, Tchikov V, and Schneider-Brachert W (2008). Regulation of TNFR1 and CD95 signalling by receptor compartmentalization. Nat Rev Mol Cell Biol 9, 655–662. 10.1038/nrm2430. [DOI] [PubMed] [Google Scholar]
- 22.Taylor MJ, Perrais D, and Merrifield CJ (2011). A high precision survey of the molecular dynamics of mammalian clathrin-mediated endocytosis. PLoS Biol 9, e1000604. 10.1371/journal.pbio.1000604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yamamoto H, Kishimoto T, and Minamoto S (1998). NF-kappaB activation in CD27 signaling: involvement of TNF receptor-associated factors in its signaling and identification of functional region of CD27. J Immunol 161, 4753–4759. [PubMed] [Google Scholar]
- 24.Choi S, Warzecha C, Zvezdova E, Lee J, Argenty J, Lesourne R, Aravind L, and Love PE (2017). THEMIS enhances TCR signaling and enables positive selection by selective inhibition of the phosphatase SHP-1. Nat Immunol 18, 433–441. 10.1038/ni.3692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chiang GG, and Sefton BM (2001). Specific dephosphorylation of the Lck tyrosine protein kinase at Tyr-394 by the SHP-1 protein-tyrosine phosphatase. J Biol Chem 276, 23173–23178. 10.1074/jbc.M101219200. [DOI] [PubMed] [Google Scholar]
- 26.Holdorf AD, Lee KH, Burack WR, Allen PM, and Shaw AS (2002). Regulation of Lck activity by CD4 and CD28 in the immunological synapse. Nat Immunol 3, 259–264. 10.1038/ni761. [DOI] [PubMed] [Google Scholar]
- 27.Lovatt M, Filby A, Parravicini V, Werlen G, Palmer E, and Zamoyska R (2006). Lck regulates the threshold of activation in primary T cells, while both Lck and Fyn contribute to the magnitude of the extracellular signal-related kinase response. Mol Cell Biol 26, 8655–8665. 10.1128/MCB.00168-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cuevas B, Lu Y, Watt S, Kumar R, Zhang J, Siminovitch KA, and Mills GB (1999). SHP-1 regulates Lck-induced phosphatidylinositol 3-kinase phosphorylation and activity. J Biol Chem 274, 27583–27589. 10.1074/jbc.274.39.27583. [DOI] [PubMed] [Google Scholar]
- 29.Colombetti S, Basso V, Mueller DL, and Mondino A (2006). Prolonged TCR/CD28 engagement drives IL-2-independent T cell clonal expansion through signaling mediated by the mammalian target of rapamycin. J Immunol 176, 2730–2738. 10.4049/jimmunol.176.5.2730. [DOI] [PubMed] [Google Scholar]
- 30.Pais Ferreira D, Silva JG, Wyss T, Fuertes Marraco SA, Scarpellino L, Charmoy M, Maas R, Siddiqui I, Tang L, Joyce JA, et al. (2020). Central memory CD8(+) T cells derive from stem-like Tcf7(hi) effector cells in the absence of cytotoxic differentiation. Immunity 53, 985–1000 e1011. 10.1016/j.immuni.2020.09.005. [DOI] [PubMed] [Google Scholar]
- 31.Chi H (2012). Regulation and function of mTOR signalling in T cell fate decisions. Nat Rev Immunol 12, 325–338. 10.1038/nri3198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sukumar M, Liu J, Ji Y, Subramanian M, Crompton JG, Yu Z, Roychoudhuri R, Palmer DC, Muranski P, Karoly ED, et al. (2013). Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J Clin Invest 123, 4479–4488. 10.1172/JCI69589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Fischer M, Bantug GR, Dimeloe S, Gubser PM, Burgener AV, Grahlert J, Balmer ML, Develioglu L, Steiner R, Unterstab G, et al. (2018). Early effector maturation of naive human CD8(+) T cells requires mitochondrial biogenesis. Eur J Immunol 48, 1632–1643. 10.1002/eji.201747443. [DOI] [PubMed] [Google Scholar]
- 34.Zhang H, Jadhav RR, Cao W, Goronzy IN, Zhao TV, Jin J, Ohtsuki S, Hu Z, Morales J, Greenleaf WJ, et al. (2022). Aging-associated HELIOS deficiency in naive CD4(+) T cells alters chromatin remodeling and promotes effector cell responses. Nat Immunol. 10.1038/s41590-022-01369-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Feng X, Wang H, Takata H, Day TJ, Willen J, and Hu H (2011). Transcription factor Foxp1 exerts essential cell-intrinsic regulation of the quiescence of naive T cells. Nat Immunol 12, 544–550. 10.1038/ni.2034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wei H, Geng J, Shi B, Liu Z, Wang YH, Stevens AC, Sprout SL, Yao M, Wang H, and Hu H (2016). Cutting Edge: Foxp1 Controls Naive CD8+ T Cell Quiescence by Simultaneously Repressing Key Pathways in Cellular Metabolism and Cell Cycle Progression. J Immunol 196, 3537–3541. 10.4049/jimmunol.1501896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhao X, Shan Q, and Xue HH (2022). TCF1 in T cell immunity: a broadened frontier. Nat Rev Immunol 22, 147–157. 10.1038/s41577-021-00563-6. [DOI] [PubMed] [Google Scholar]
- 38.Wu N, Zheng B, Shaywitz A, Dagon Y, Tower C, Bellinger G, Shen CH, Wen J, Asara J, McGraw TE, et al. (2013). AMPK-dependent degradation of TXNIP upon energy stress leads to enhanced glucose uptake via GLUT1. Mol Cell 49, 1167–1175. 10.1016/j.molcel.2013.01.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kanbar JN, Ma S, Kim ES, Kurd NS, Tsai MS, Tysl T, Widjaja CE, Limary AE, Yee B, He Z, et al. (2022). The long noncoding RNA Malat1 regulates CD8+ T cell differentiation by mediating epigenetic repression. J Exp Med 219. 10.1084/jem.20211756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ramana CV, Chatterjee-Kishore M, Nguyen H, and Stark GR (2000). Complex roles of Stat1 in regulating gene expression. Oncogene 19, 2619–2627. 10.1038/sj.onc.1203525. [DOI] [PubMed] [Google Scholar]
- 41.Ohmori Y, Schreiber RD, and Hamilton TA (1997). Synergy between interferon-gamma and tumor necrosis factor-alpha in transcriptional activation is mediated by cooperation between signal transducer and activator of transcription 1 and nuclear factor kappaB. J Biol Chem 272, 14899–14907. 10.1074/jbc.272.23.14899. [DOI] [PubMed] [Google Scholar]
- 42.Kye YC, Lee GW, Lee SW, Ju YJ, Kim HO, Yun CH, and Cho JH (2021). STAT1 maintains naive CD8(+) T cell quiescence by suppressing the type I IFN-STAT4-mTORC1 signaling axis. Sci Adv 7, eabg8764. 10.1126/sciadv.abg8764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chiang EY, de Almeida PE, de Almeida Nagata DE, Bowles KH, Du X, Chitre AS, Banta KL, Kwon Y, McKenzie B, Mittman S, et al. (2020). CD96 functions as a costimulatory receptor to enhance CD8(+) T cell activation and effector responses. Eur J Immunol 50, 891–902. 10.1002/eji.201948405. [DOI] [PubMed] [Google Scholar]
- 44.Kaech SM, and Cui W (2012). Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol 12, 749–761. 10.1038/nri3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Watson G, Ronai ZA, and Lau E (2017). ATF2, a paradigm of the multifaceted regulation of transcription factors in biology and disease. Pharmacol Res 119, 347–357. 10.1016/j.phrs.2017.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kim MV, Ouyang W, Liao W, Zhang MQ, and Li MO (2013). The transcription factor Foxo1 controls central-memory CD8+ T cell responses to infection. Immunity 39, 286–297. 10.1016/j.immuni.2013.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.He B, Xing S, Chen C, Gao P, Teng L, Shan Q, Gullicksrud JA, Martin MD, Yu S, Harty JT, et al. (2016). CD8(+) T Cells Utilize Highly Dynamic Enhancer Repertoires and Regulatory Circuitry in Response to Infections. Immunity 45, 1341–1354. 10.1016/j.immuni.2016.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tesselaar K, Arens R, van Schijndel GM, Baars PA, van der Valk MA, Borst J, van Oers MH, and van Lier RA (2003). Lethal T cell immunodeficiency induced by chronic costimulation via CD27-CD70 interactions. Nat Immunol 4, 49–54. 10.1038/ni869. [DOI] [PubMed] [Google Scholar]
- 49.Zapata JM, Perez-Chacon G, Carr-Baena P, Martinez-Forero I, Azpilikueta A, Otano I, and Melero I (2018). CD137 (4–1BB) Signalosome: Complexity Is a Matter of TRAFs. Front Immunol 9, 2618. 10.3389/fimmu.2018.02618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, and Vale RD (2017). T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433. 10.1126/science.aaf1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kretschmer L, Flossdorf M, Mir J, Cho YL, Plambeck M, Treise I, Toska A, Heinzel S, Schiemann M, Busch DH, and Buchholz VR (2020). Differential expansion of T central memory precursor and effector subsets is regulated by division speed. Nat Commun 11, 113. 10.1038/s41467-019-13788-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bresser K, Kok L, Swain AC, King LA, Jacobs L, Weber TS, Perie L, Duffy KR, de Boer RJ, Scheeren FA, and Schumacher TN (2022). Replicative history marks transcriptional and functional disparity in the CD8(+) T cell memory pool. Nat Immunol 23, 791–801. 10.1038/s41590-022-01171-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wolf T, Jin W, Zoppi G, Vogel IA, Akhmedov M, Bleck CKE, Beltraminelli T, Rieckmann JC, Ramirez NJ, Benevento M, et al. (2020). Dynamics in protein translation sustaining T cell preparedness. Nat Immunol 21, 927–937. 10.1038/s41590-020-0714-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Delpoux A, Marcel N, Hess Michelini R, Katayama CD, Allison KA, Glass CK, Quinones-Parra SM, Murre C, Loh L, Kedzierska K, et al. (2021). FOXO1 constrains activation and regulates senescence in CD8 T cells. Cell Rep 34, 108674. 10.1016/j.celrep.2020.108674. [DOI] [PubMed] [Google Scholar]
- 55.Coquet JM, Ribot JC, Babala N, Middendorp S, van der Horst G, Xiao Y, Neves JF, Fonseca-Pereira D, Jacobs H, Pennington DJ, et al. (2013). Epithelial and dendritic cells in the thymic medulla promote CD4+Foxp3+ regulatory T cell development via the CD27-CD70 pathway. J Exp Med 210, 715–728. 10.1084/jem.20112061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Konopacki C, Pritykin Y, Rubtsov Y, Leslie CS, and Rudensky AY (2019). Transcription factor Foxp1 regulates Foxp3 chromatin binding and coordinates regulatory T cell function. Nat Immunol 20, 232–242. 10.1038/s41590-018-0291-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bandaranayake AD, Correnti C, Ryu BY, Brault M, Strong RK, and Rawlings DJ (2011). Daedalus: a robust, turnkey platform for rapid production of decigram quantities of active recombinant proteins in human cell lines using novel lentiviral vectors. Nucleic Acids Res 39, e143. 10.1093/nar/gkr706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Mirdita M, Schutze K, Moriwaki Y, Heo L, Ovchinnikov S, and Steinegger M (2022). ColabFold: making protein folding accessible to all. Nat Methods 19, 679–682. 10.1038/s41592-022-01488-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Goddard TD, Huang CC, Meng EC, Pettersen EF, Couch GS, Morris JH, and Ferrin TE (2018). UCSF ChimeraX: Meeting modern challenges in visualization and analysis. Protein Sci 27, 14–25. 10.1002/pro.3235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Nicholson IC, Lenton KA, Little DJ, Decorso T, Lee FT, Scott AM, Zola H, and Hohmann AW (1997). Construction and characterisation of a functional CD19 specific single chain Fv fragment for immunotherapy of B lineage leukaemia and lymphoma. Mol Immunol 34, 1157–1165. 10.1016/s0161-5890(97)00144-2. [DOI] [PubMed] [Google Scholar]
- 61.Yang J, Baskar S, Kwong KY, Kennedy MG, Wiestner A, and Rader C (2011). Therapeutic potential and challenges of targeting receptor tyrosine kinase ROR1 with monoclonal antibodies in B-cell malignancies. PLoS One 6, e21018. 10.1371/journal.pone.0021018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Davila ML, Kloss CC, Gunset G, and Sadelain M (2013). CD19 CAR-targeted T cells induce long-term remission and B Cell Aplasia in an immunocompetent mouse model of B cell acute lymphoblastic leukemia. PLoS One 8, e61338. 10.1371/journal.pone.0061338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Paszkiewicz PJ, Frassle SP, Srivastava S, Sommermeyer D, Hudecek M, Drexler I, Sadelain M, Liu L, Jensen MC, Riddell SR, and Busch DH (2016). Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia. J Clin Invest 126, 4262–4272. 10.1172/JCI84813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Inaba K, Inaba M, Romani N, Aya H, Deguchi M, Ikehara S, Muramatsu S, and Steinman RM (1992). Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med 176, 1693–1702. 10.1084/jem.176.6.1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bullock TN, and Yagita H (2005). Induction of CD70 on dendritic cells through CD40 or TLR stimulation contributes to the development of CD8+ T cell responses in the absence of CD4+ T cells. J Immunol 174, 710–717. 10.4049/jimmunol.174.2.710. [DOI] [PubMed] [Google Scholar]
- 66.van der Windt GJW, Chang CH, and Pearce EL (2016). Measuring Bioenergetics in T Cells Using a Seahorse Extracellular Flux Analyzer. Curr Protoc Immunol 113, 3 16B 11–13 16B 14. 10.1002/0471142735.im0316bs113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Feng J, Liu T, Qin B, Zhang Y, and Liu XS (2012). Identifying ChIP-seq enrichment using MACS. Nat Protoc 7, 1728–1740. 10.1038/nprot.2012.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Stuart T, Srivastava A, Madad S, Lareau CA, and Satija R (2021). Single-cell chromatin state analysis with Signac. Nat Methods 18, 1333–1341. 10.1038/s41592-021-01282-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hafemeister C, and Satija R (2019). Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol 20, 296. 10.1186/s13059-019-1874-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, Baglaenko Y, Brenner M, Loh PR, and Raychaudhuri S (2019). Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods 16, 1289–1296. 10.1038/s41592-019-0619-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hao Y, Hao S, Andersen-Nissen E, Mauck WM 3rd, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, et al. (2021). Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 e3529. 10.1016/j.cell.2021.04.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Schep AN, Wu B, Buenrostro JD, and Greenleaf WJ (2017). chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat Methods 14, 975–978. 10.1038/nmeth.4401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kartha VK, Duarte FM, Hu Y, Ma S, Chew JG, Lareau CA, Earl A, Burkett ZD, Kohlway AS, Lebofsky R, and Buenrostro JD (2022). Functional inference of gene regulation using single-cell multi-omics. Cell Genom 2. 10.1016/j.xgen.2022.100166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M, Lennon NJ, Livak KJ, Mikkelsen TS, and Rinn JL (2014). The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 32, 381–386. 10.1038/nbt.2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Multiome-seq data have been deposited at the GEO repository and are publicly available as of the date of publication. Accession number is listed in the key resources table. This paper does not report original code. Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
Key resources table.
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Antibodies | ||
| THE™ HIS Tag Antibody – iFluor 647 (1:200) | GenScript | Cat#A01802; RRID: AB_2943644 |
| Brilliant Violet 785 anti-human CD45 (clone HI30, 1:100) | BioLegend | Cat#304031; RRID: AB_10900423 |
| BD Horizon BV421 anti-human CD45RO (clone UCHL1, 1:100) | BD Biosciences | Cat#562641; RRID: AB_2737696 |
| BD Horizon BUV395 anti-human CD45RO (clone UCHL1, 1:40) | BD Biosciences | Cat#564292; RRID: AB_2744410 |
| Alexa Fluor 700 anti-human CD45RA (clone HI100, 1:100) | BioLegend | Cat#304119; RRID: AB_493762 |
| PerCP/Cyanine5.5 anti-human CCR7 (clone G043H7, 1:100) | BioLegend | Cat#353219; RRID: AB_10915275 |
| Brilliant Violet 785 anti-mouse/rat/human CD27 (clone LG3.A10, 1:800) | BioLegend | Cat#124241; RRID: AB_2800595 |
| Brilliant Violet 510 anti-mouse/rat/human CD27 (clone LG3.A10, 1:200) | BioLegend | Cat#124229; RRID: AB_2565795 |
| APC/Cyanine7 anti-human CD27 (clone MT-271, 1:100) | BioLegend | Cat#356423; RRID: AB_2566772 |
| FITC anti-human CD27 (clone O323, 1:100) | BioLegend | Cat#302805; RRID: AB_314297 |
| PerCP/Cyanine5.5 anti-human CD70 (clone 113-16, 1:100) | BioLegend | Cat#355107; RRID: AB_2562478 |
| eFluor 450 anti-human CD69 Monoclonal Antibody (clone FN50, 1:100) | eBioscience | Cat#48-0699-42; RRID: AB_2574025 |
| PE/Cyanine7 anti-human CD137 (clone 4B4-1, 1:100) | BioLegend | Cat#309817; RRID: AB_2287731 |
| BD OptiBuild BUV737 mouse anti-human CD137 (clone 4B4-1, 1:100) | BD Biosciences | Cat#741861; RRID: AB_2871191 |
| APC anti-human CD25 (clone BC96, 1:200) | BioLegend | Cat#302609; RRID: AB_314279 |
| Pacific Blue anti-human CD127 (clone A019D5, 1:50) | BioLegend | Cat#351305; RRID: AB_10718638 |
| Brilliant Violet 605 anti-human CD95 (clone DX2, 1:100) | BioLegend | Cat#305627; RRID: AB_2562444 |
| FITC anti-human CD8α (clone RPA-T8, 1:500) | BioLegend | Cat#301005; RRID: AB_314123 |
| BD Horizon BUV496 mouse anti-human CD8 (clone RPA-T8, 1:500) | BD Biosciences | Cat#612943; RRID: AB_2916884 |
| PE anti-human CD279 (PD-1) Monoclonal Antibody (clone eBioJ105, 1:100) | eBioscience | Cat#12-2799-42; RRID: AB_11042478 |
| PE-Cyanine7 anti-human CD279 (PD-1) Monoclonal Antibody (clone eBioJ105, 1:100) | eBioscience | Cat#25-2799-42; RRID: AB_10853804 |
| BD Horizon PE-CF594 mouse anti-human TIM-3 (clone 7D3, 1:100) | BD Biosciences | Cat#565561; RRID: AB_2744371 |
| Alexa Fluor 647 anti-human TIGIT (clone A15153G, 1:100) | BioLegend | Cat#372723; RRID: AB_2715971 |
| Brilliant Violet 605 anti-human CD223 (LAG-3) (clone 11C3C65, 1:100) | BioLegend | Cat#369324; RRID: AB_2721541 |
| Brilliant Violet 711 anti-human CD39 (clone A1, 1:100) | BioLegend | Cat#328227; RRID: AB_2632893 |
| PE anti-human EGFR (clone AY13, 1:100) | BioLegend | Cat#352903; RRID: AB_10898161 |
| APC anti-human EGFR (clone AY13, 1:100) | BioLegend | Cat#352905; RRID: AB_11148943 |
| APC-Cyanine7 anti-human CD62L (clone DREG-56, 1:100) | BioLegend | Cat#304813; RRID: AB_493583 |
| FITC anti-human CD19 (clone HIB19, 1:100) | BioLegend | Cat#302205; RRID: AB_314235 |
| PE anti-human CD19 (clone HIB19, 1:100) | BioLegend | Cat#302207; RRID: AB_314237 |
| BD Horizon BV421 anti-human Granzyme B (clone GB11, 1:100) | BD Biosciences | Cat#563389; RRID: AB_2738175 |
| PE mouse anti-TCF-7/TCF-1 (clone S33-966, 1:100) | BD Biosciences | Cat#564217; RRID: AB_2687845 |
| PE mouse IgG1k Isotype Control | BD Biosciences | Cat#554680; RRID: AB_395506 |
| APC anti-human IL-2 (clone MQ1-17H12, 1:100) | eBioscience | Cat#17-7029-82; RRID: AB_469492 |
| PE-Cyanine7 anti-human TNF-α (clone Mab11, 1:100) | BioLegend | Cat#502929; RRID: AB_2204080 |
| PE anti-human IFN gamma (clone 4S.B3, 1:100) | eBioscience | Cat#12-7319-42; RRID: AB_1311247 |
| PE-Cy7 anti-mouse Ki-67 (clone B56, 1:100) | BD Biosciences | Cat#561283; RRID: AB_10716060 |
| PE-Cyanine7 anti-mouse CD39 (clone Duha59, 1:100) | BioLegend | Cat#143805; RRID: AB_2563393 |
| Alexa Fluor 700 anti-mouse CD45.1 (clone A20, 1:200) | BioLegend | Cat#110723; RRID: AB_493732 |
| Brilliant Violet 605 anti-mouse CD279 (PD-1) (clone 29F.1A12, 1:100) | BioLegend | Cat#135219; RRID: AB_11125371 |
| BV711 mouse anti-mouse TIGIT (clone 1G9, 1:100) | BD Biosciences | Cat#744214; RRID: AB_2742063 |
| Brilliant Violet 421 anti-mouse CD366 (TIM-3) (clone RMT3-23, 1:100) | BioLegend | Cat#119723; RRID: AB_2616908 |
| BUV661 anti-mouse CD19 (clone 1D3, 1:200) | BD Biosciences | Cat#612971; RRID: AB_2870243 |
| Brilliant Violet 785 anti-mouse CD223 (LAG-3) (clone C9B7W, 1:100) | BioLegend | Cat#125219; RRID: AB_2566571 |
| BUV395 rat anti-mouse CD8a (clone 53-6.7, 1:500) | BD Biosciences | Cat#565968; RRID: AB_2739421 |
| Pacific Blue rat anti-mouse CD8a (clone 53-6.7, 1:500) | BD Biosciences | Cat#558106; RRID: AB_397029 |
| BUV496 rat anti-mouse CD4 (clone GK1.5, 1:200) | BD Biosciences | Cat#612952; RRID: AB_2813886 |
| PE/Cyanine7 anti-mouse CD70 (clone FR70, 1:300) | BioLegend | Cat#104611; RRID: AB_2750466 |
| FITC anti-mouse CD11c (clone N418, 1:300) | BioLegend | Cat#117305; RRID: AB_313774 |
| BV421 rat anti-mouse I-A/I-E (clone M5/114.15.2, 1:400) | BD Biosciences | Cat#562564; RRID: AB_2716857 |
| APC anti-mouse CD80 (clone 16-10AL, 1:300) | BioLegend | Cat#104713; RRID: AB_313134 |
| Alexa Fluor 700 anti-mouse CD86 (clone GL-1, 1:200) | BioLegend | Cat#105023; RRID: AB_493720 |
| BV711 mouse Anti-Ki-67 (clone B56, 1:100) | BD Biosiences | Cat#563755; RRID: AB_2738406 |
| PE anti-human ERK1/2 phospho [T202,Y204] (clone 6B8B69, 1:30) | BioLegend | Cat#369506; RRID: AB_2629705 |
| Alexa Fluor 488 anti-human Akt phospho [Thr308] (clone D25E6, 1:50) | Cell Signaling Technology | Cat#43506; RRID: AB_2799244 |
| Alexa Fluor 488 anti-human SHP-1 Rabbit mAb (clone E1U6R, 1:2000) | Cell Signaling Technology | Cat#77181; RRID: AB_2943643 |
| Alexa Fluor 488 Rabbit mAb IgG XP Isotype Control (clone DA1E, 1:1000) | Cell Signaling Technology | Cat#2975; RRID: AB_10699151 |
| Pacific Blue mouse anti-human CD8 (clone RPA-T8, 1:100) | BD Biosciences | Cat#558207; RRID: AB_397058 |
| InVivoMAb anti-mouse CD70 | BioXCell | Cat#BE0022; RRID: AB_1107667 |
| InVivoMAb rat IgG2b isotype control | BioXCell | Cat#BE0090; RRID: AB_1107780 |
| TotalSeq™-A0251 anti-human Hashtag 1 Antibody | BioLegend | Cat#394601; RRID: AB_2750015 |
| TotalSeq™-A0252 anti-human Hashtag 2 Antibody | BioLegend | Cat#394603; RRID: AB_2750016 |
| TotalSeq™-A0253 anti-human Hashtag 3 Antibody | BioLegend | Cat#394605; RRID: AB_2750017 |
| TotalSeq™-A0254 anti-human Hashtag 4 Antibody | BioLegend | Cat#394607; RRID: AB_2750018 |
| TotalSeq™-A0255 anti-human Hashtag 5 Antibody | BioLegend | Cat#394609; RRID: AB_2750019 |
| TotalSeq™-A0256 anti-human Hashtag 6 Antibody | BioLegend | Cat#394611; RRID: AB_2750020 |
| TotalSeq™-A0257 anti-human Hashtag 7 Antibody | BioLegend | Cat#394613; RRID: AB_2750021 |
| TotalSeq™-A0258 anti-human Hashtag 8 Antibody | BioLegend | Cat#394615; RRID: AB_2750022 |
| Anti-human CD27 (clone E6S8H) Rabbit mAb | Cell Signaling Technology | Cat#28034; RRID: AB_2943642 |
| Anti-human β2-microgloblulin (clone D8P1H) Rabbit mAb | Cell Signaling Technology | Cat#12851; RRID: AB_2716551 |
| Anti-human β–Actin (clone 13E5) Rabbit mAb | Cell Signaling Technology | Cat#4770; RRID: AB_2223172 |
| Anti-human GAPDH (clone D16H11) XP Rabbit mAb | Cell Signaling Technology | Cat#5174; RRID: AB_10622025 |
| Anti-human TRAF2 (clone C192) Rabbit mAb | Cell Signaling Technology | Cat#4724; RRID: AB_2209845 |
| Anti-human SHP-1 (clone E1U6R) Rabbit mAb | Cell Signaling Technology | Cat#26516; RRID: AB_2934293 |
| Anti-human SHP-1 phospho [Tyr564] (clone D11G5) Rabbit mAb | Cell Signaling Technology | Cat#8849; RRID: AB_11141050 |
| Anti-human SH-PTP-2 (clone B-1) Mouse mAb | Santa Cruz Biotechnology | Cat#sc-7384; RRID: AB_628252 |
| Anti-human SHP1 phospho [Tyr536] | Abcam | Cat#ab41437; RRID: AB_777914 |
| Anti-human SHP1 phospho [Tyr591] | Abcam | Cat#ab41436; RRID: AB_777913 |
| Anti-human Lck (clone D88) XP Rabbit mAb | Cell Signaling Technology | Cat#2984; RRID: AB_2136313 |
| Anti-human phospho Lck [Tyr394] (clone E5L3D) Rabbit mAb | Cell Signaling Technology | Cat#70926; RRID: AB_2924371 |
| Anti-human phospho Lck [Tyr505] | Cell Signaling Technology | Cat#2751; RRID: AB_330446 |
| Anti-mouse IgG, HRP-linked Antibody | Cell Signaling Technology | Cat#7076; RRID: AB_330924 |
| Anti-rabbit IgG, HRP-linked Antibody | Cell Signaling Technology | Cat#7074; RRID: AB_2099233 |
| Bacterial and virus strains | ||
| Turbo Competent E. coli (MAX Efficiency DH5α) | Invitrogen | Cat#18258-012 |
| Biological samples | ||
| Human CD8+ T cells isolated from PBMCs | Fred Hutchinson Cancer Center / Bloodworks Northwest | N/A |
| Chemicals, peptides, and recombinant proteins | ||
| ES91-17, 100 μM | Sigma-Aldrich | Cat#SML2712; CAS: 55854-43-8 |
| Mdivi-1, 50 μM | Selleck | Cat#S7162; CAS: 338967-87-6 |
| Latrunculin A, 0.04 μM | Sigma-Aldrich | Cat#76343-93-6; CAS: 76343-93-6 |
| Dynasore, see titration | Selleck | Cat#S8047; CAS: 304448-55-3 |
| Dyngo-4a, see titration | Selleck | Cat#S7163; CAS: 1256493-34-1 |
| ULK-101, 5 μM | Selleck | Cat#S8793; CAS: 2443816-45-1 |
| Lactacystin, 20 μM | Tocris | Cat#2267; CAS: 133343-34-7 |
| TPI-1, 0.4 μM | Selleck | Cat#S6570; CAS: 79756-69-7 |
| GM6001, 10 μM | Selleck | Cat#S7157; CAS: 142880-36-2 |
| MitoTracker™ Green, 100 nM | ThermoFisher | Cat#M7514 |
| CellTrace™ CFSE dye, 0.1 μM | ThermoFisher | Cat#C34554 |
| 7-AAD | Sigma-Aldrich | Cat#SML1633 |
| Recombinant human IL-2 (clinical grade Proleukin®, Aldesleukin) | Prometheus Laboratories | N/A |
| Recombinant human IL-7 | PeproTech | Cat#200-07; GenPept: P13232 |
| Recombinant mouse GM-CSF | PeproTech | Cat#315-03; GenPept: P01587 |
| Ultra-LEAF™ Purified anti-human CD3 (clone OKT3) | BioLegend | Cat#317347 RRID: AB_2571994 |
| Anti-human CD28 (clone TGN1412) | This paper | N/A |
| Ultra-LEAF™ Purified anti-human CD28 (clone CD28.2) | BioLegend | Cat#302934 RRID: AB_11148949 |
| Anti-human CD70 Dimer Trimer (CD70DT) | This paper | N/A |
| IgG1-Fc | This paper | N/A |
| Purified NA/LE Hamster Anti-Mouse CD28 (clone 37.51) | BD Pharmingen | Cat#553294 RRID: AB_394763 |
| Purified NA/LE Hamster Anti-Mouse CD3ε (clone 145-2C11) | BD Pharmingen | Cat#553057 RRID: AB_394590 |
| Dynabeads™ Human T-Activator CD3/CD28 | Gibco | Cat#11131D |
| Critical commercial assays | ||
| Chromium Next GEM Single Cell Multiome ATAC + Gene Expression Reagent Bundle | 10X Genomics | Cat#1000258 |
| Chromium Next GEM Chip J Single Cell Kit | 10X Genomics | Cat#1000230 |
| Chromium Next GEM Chip G Single Cell Kit | 10X Genomics | Cat#1000127 |
| CalPhos Mammalian Transfection Kit | Clonetech / Takara | Cat#631312 |
| Seahorse XF Glycolysis Stress Test Kit | Agilent | Cat#103020-100 |
| Seahorse XF Mitochondrial Stress Test Kit | Agilent | Cat#103015-100 |
| P3 Primary Cell 4D-Nucleofector™ X Kit S | Lonza | Cat#V4XP-3032 |
| NucleoSpin RNA XS Micro kit for RNA purification | Macherey-Nagel | Cat#740902 |
| SuperScript IV First-Strand Synthesis System Kit | ThermoFisher | Cat#18091050 |
| EasySep™ Human Naïve CD8+ T Cell Isolation Kit II | STEMCELL | Cat#17968 |
| EasySep™ Human CD8+ T Cell Isolation Kit | STEMCELL | Cat#17953 |
| EasySep™ Mouse CD8+ T Cell Isolation Kit | STEMCELL | Cat#19853 |
| Pierce™ BCA Protein Assay Kit | ThermoFisher | Cat#23225 |
| Dynabeads™ Protein G Immunoprecipitation Kit | Invitrogen | Cat#10007D |
| Deposited data | ||
| Multiome Single Cell RNAseq data | This paper | GEO: GSE244834 |
| Experimental models: Cell lines | ||
| Lenti-X™ 293T cells | Clonetech / Takara | Cat#632180 RRID: CVCL_4401 |
| K32 | Carl June, Univerisy of Pennsylvania (Maus et al., 2002) | N/A |
| CD70-transgene expressing K32 | This paper | N/A |
| Raji | ATCC | Cat#CCL-86 RRID: CVCL_0511 |
| Raji+ffluc+GFP | This paper | N/A |
| MDA-MB-231+ffluc+GFP | This paper | N/A |
| NCI-H1703 | ATCC | Cat#CRL-5889 RRID: CVCL_1490 |
| NIH-3T3 | ATCC | Cat#CRL-1658 RRID: CVCL_0594 |
| CD40L-transgene expressing NIH-3T3 | This paper | N/A |
| Platinum-E cells | Cell Biolabs Inc. | Cat#RV-101 RRID: CVCL_B488 |
| Experimental models: Organisms/strains | ||
| Mouse: C57BL/6 wildtype (WT) | The Jackson Laboratory | RRID: IMSR_JAX:000664 |
| Mouse: B6.SJL-Ptprca Pepcb/BoyJ (CD45.1) | The Jackson Laboratory | RRID: IMSR_JAX:002014 |
| Mouse: C57BL/6-Tg(CAG-OVAL)916Jen/J (Act-mOVA) | The Jackson Laboratory | RRID: IMSR_JAX:005145 |
| Mouse: C57BL/6-Tg(TcraTcrb)1100Mjb/J (OT-1) | The Jackson Laboratory | RRID: IMSR_JAX:003831 |
| Mouse: NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) | The Jackson Laboratory | RRID: IMSR_JAX:005557 |
| Oligonucleotides | ||
| sgRNA targeting sequence: SHP-1 (PTPN6): 5’ [CAUCCAGCCCACUGAGGUCU] 3’ | Synthego | N/A |
| Primer: PTPN6_Ex2_Forward1: 5’ [CTCTCTGCCTGCCCAGACTA] 3’ | IDT | N/A |
| Primer: PTPN6_Ex2_Reverse1: 5’ [ACGGGGAACCAGGAATGAGTG] 3’ | IDT | N/A |
| TaqMan Gene Expression Assay: B2M (Hs00187842_m1) | ThermoFisher Scientific | Cat#4331182 |
| TaqMan Gene Expression Assay: CD27 (Hs00386811_m1) | ThermoFisher Scientific | Cat#4331182 |
| Recombinant DNA | ||
| Lentiviral Packaging Vector: pCHGP-2 | This paper | N/A |
| Lentiviral Packaging Vector: pCMV-Rev2 | This paper | N/A |
| Lentiviral Packaging Vector: pCMV-G | This paper | N/A |
| Plasmid: epHIV7_hCD19(FMC63)_IgG4_CD28TMD_h41BB_ hCD3ζ_T2A_EGFRt | This paper | N/A |
| Plasmid: epHIV7_hROR1(R12)_IgG4_CD28TMD_ h41BB_ hCD3ζ_T2A_CD19t | This paper | N/A |
| Plasmid: epHIV7_hROR1(R12)_IgG4_CD28TMD_ h41BB_ hCD3ζ_T2A_EGFRt | This paper | N/A |
| Plasmid: epMP71-mCD19(1D3)_IgG4D28TMD_m41BB_ mCD3ζ_T2A_mCD19t | This paper | N/A |
| Plasmid: epHIV7_hCD70 | This paper | N/A |
| Plasmid: ep-HIV7_mCD40L | This paper | N/A |
| Software and algorithms | ||
| FlowJo software v10.8.1 | Becton Dickinson & Company | https://www.flowjo.com/ |
| Thermo TraceFinder 4.1 | Thermo Fisher Scientific | https://www.thermofisher.com/us/en/home/industrial/mass-spectrometry/liquid-chromatography-mass-spectrometry-lc-ms/lc-ms-software/lc-ms-data-acquisition-software/tracefinder-software.html |
| FACSDIVA v9.1 | Becton Dickinson & Company | https://www.bdbiosciences.com/en-us/products/software/instrumentsoftware/bd-facsdiva-software |
| ID A v2.0 Analysis Software | SONY | N/A |
| Prism v9.4.0 | GraphPad | https://www.graphpad.com/features |
| Living Image Analysis Software | Perkin Elmer | http://www.perkinelmer.com/product/li-software-for-spectrum-1-seat-addon-128113 |
| R v4.2.0 | CRAN | https://cran.r-project.org/mirrors.html |
| RStudio v2022.02.3 | RStudio | https://rstudio.com/ |
| Cellranger ARC | 10X Genomics | https://support.10xgenomics.com/single-cell-multiome-atac-gex/software/downloads |
| Seurat | (Stuart et al., 2021) | https://github.com/satijalab/seurat |
| MACS2 | (Feng et al., 2012) | https://pypi.org/project/MACS2/ |
| Signac | (Stuart et al., 2021) | https://stuartlab.org/signac/ |
| Harmony | (Korsunsky et al., 2019) | https://portals.broadinstitute.org/harmony/index.html |
| ChromVAR | (Schep et al., 2017) | https://greenleaflab.github.io/chromVAR/index.html |
| FigR | (Kartha et al., 2022) | https://buenrostrolab.github.io/FigR/ |
| Monocle3 | (Trapnell et al., 2014) | https://cole-trapnell-lab.github.io/monocle3/ |
| Other | ||
| Trans-blot Turbo System | Bio-Rad | N/A |
| PVDF membranes | Bio-Rad | Cat#10026934 |
| Mini-PROTEAN® TGX™ Precast Protein Gels | Bio-Rad | Cat#456-1085 |
| ECL Chemiluminescent Substrate | Bio-Rad | Cat#1705062 |
