16
Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019 SCIENCE IMMUNOLOGY | RESEARCH ARTICLE 1 of 15 T CELLS Modulation of asymmetric cell division as a mechanism to boost CD8 + T cell memory Mariana Borsa 1 , Isabel Barnstorf 1 *, Nicolas S. Baumann 1 *, Katharina Pallmer 1 *, Alexander Yermanos 1 , Fabienne Gräbnitz 1 , Niculò Barandun 1 , Annika Hausmann 1 , Ioana Sandu 1 , Yves Barral 2 , Annette Oxenius 1† Asymmetric partitioning of fate determinants is a mechanism that contributes to T cell differentiation. However, it remains unclear whether the ability of T cells to divide asymmetrically is influenced by their differentiation state, as well as whether enforcing asymmetric cell division (ACD) rates would have an impact on T cell differentiation and memory formation. Using the murine LCMV infection model, we established a correlation between cell stem- ness and the ability of CD8 + T cells to undergo ACD. Transient mTOR inhibition was proven to increase ACD rates in naïve and memory cells and to install this ability in exhausted CD8 + T cells. Functionally, enforced ACD correlated with increased memory potential, leading to more efficient recall response and viral control upon acute or chronic LCMV infection. Moreover, transient mTOR inhibition also increased ACD rates in human CD8 + T cells. Transcrip- tional profiling revealed that progenies emerging from enforced ACD exhibited more pronounced early memory signatures, which functionally endowed these cells with better survival in the absence of antigen exposure and more robust homing to secondary lymphoid organs, providing critical access to survival niches. Our data provide important insights into how ACD can improve long-term survival and function of T cells and open new perspectives for vaccination and adoptive T cell transfer therapies. INTRODUCTION CD8 + T cells play a key role in adaptive immunity and contribute to host defense and antitumor responses. After initial activation, CD8 + T cells proliferate and differentiate, acquiring effector functions. After clearance of the infection, most of these effector cells undergo apoptosis during the contraction phase, but a subset of memory precursors survives and differentiates into memory cells, which are long-lived, self-renewing cells that afford increased protection against secondary encounter with the pathogen (1). How differenti- ation into effector and memory cells occurs is still incompletely un- derstood, although the contribution of specific transcription factors (2), metabolic profiles (37), epigenetic changes during the course of immune responses (89), and asymmetric cell division (ACD) (1014) has been explored. The contact interface between the antigen-presenting cell (APC) and the engaged T cell is termed immunological synapse. By estab- lishing a stable immunological synapse, T cells undergo an extensive cytoskeleton remodeling to build a global polarization axis (1516), leading to the asymmetric partitioning of surface molecules, such as the T cell receptor (TCR), CD4/CD8, cytokine receptors, cell fate determinants such as transcription factors, and metabolism such as mTOR (mammalian target of rapamycin) activity (1314). The two emerging progenies exhibit different fate potentials (11) and transcrip- tional profiles (1017), with the proximal daughter adopting an effector cell fate and the distal daughter being destined to become a memory cell (1018). Most studies analyzing ACD have concentrated on naïve murine T cells undergoing their first mitosis after TCR engagement. However, with the exception of memory CD8 + T cells, which were also reported to undergo ACD (19), it is largely unknown whether other, more terminally differentiated, antigen-experienced T cell subsets, such as effector or exhausted CD8 + T cells, can undergo ACD. Further- more, it also remains to be established whether ACD can be modulated with any functional consequence for T cell fate determination. In this study, we report that ACD is an ability linked to cell stem- ness, as only naïve and memory CD8 + T cells could divide asymmetri- cally, whereas terminally differentiated effector cells and exhausted cells, which are unable to form a self-renewing reservoir, divided sym- metrically. We identified a strategy to enforce ACD by transient in- hibition of mTOR by rapamycin. Progenies emerging from enforced ACD conditions exhibited increased memory potential because they showed improved survival and expansion upon adoptive transfer, resulting in more efficient protection upon viral challenge. Further- more, transcriptional profiling of progenies derived from symmetric and asymmetric divisions revealed strengthened memory signatures in “memory daughters” emerging from enforced ACD. Functionally, the early memory signature endowed these cells with improved sur- vival and homing to secondary lymphoid organs. Together, these data define ACD as a key mechanism for differentiation and establish- ment of a memory reservoir in murine and human CD8 + T cells. RESULTS ACD is a feature of CD8 + T cells that retain stemness As previous reports about ACD in lymphocytes focused on naïve and memory T cells, we investigated whether CD8 + T cells in other differ- entiation stages would also divide asymmetrically. To this end, we adoptively transferred naïve TCR transgenic P14 CD8 + T cells, which spe- cifically recognize the gp 33–41 peptide from the lymphocytic chorio- meningitis virus (LCMV) glycoprotein, followed by acute or chronic LCMV infection to generate different CD8 + T cell subsets. Effector cells were isolated 8 days after LCMV WE infection, memory cells were isolated at least 30 days after LCMV WE infection, and exhausted cells 1 Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland. 2 Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland. *These authors contributed equally to this work. †Corresponding author. Email: [email protected] Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works by guest on May 23, 2021 http://immunology.sciencemag.org/ Downloaded from

T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

1 of 15

T C E L L S

Modulation of asymmetric cell division as a mechanism to boost CD8+ T cell memoryMariana Borsa1, Isabel Barnstorf1*, Nicolas S. Baumann1*, Katharina Pallmer1*, Alexander Yermanos1, Fabienne Gräbnitz1, Niculò Barandun1, Annika Hausmann1, Ioana Sandu1, Yves Barral2, Annette Oxenius1†

Asymmetric partitioning of fate determinants is a mechanism that contributes to T cell differentiation. However, it remains unclear whether the ability of T cells to divide asymmetrically is influenced by their differentiation state, as well as whether enforcing asymmetric cell division (ACD) rates would have an impact on T cell differentiation and memory formation. Using the murine LCMV infection model, we established a correlation between cell stem-ness and the ability of CD8+ T cells to undergo ACD. Transient mTOR inhibition was proven to increase ACD rates in naïve and memory cells and to install this ability in exhausted CD8+ T cells. Functionally, enforced ACD correlated with increased memory potential, leading to more efficient recall response and viral control upon acute or chronic LCMV infection. Moreover, transient mTOR inhibition also increased ACD rates in human CD8+ T cells. Transcrip-tional profiling revealed that progenies emerging from enforced ACD exhibited more pronounced early memory signatures, which functionally endowed these cells with better survival in the absence of antigen exposure and more robust homing to secondary lymphoid organs, providing critical access to survival niches. Our data provide important insights into how ACD can improve long-term survival and function of T cells and open new perspectives for vaccination and adoptive T cell transfer therapies.

INTRODUCTIONCD8+ T cells play a key role in adaptive immunity and contribute to host defense and antitumor responses. After initial activation, CD8+ T cells proliferate and differentiate, acquiring effector functions. After clearance of the infection, most of these effector cells undergo apoptosis during the contraction phase, but a subset of memory precursors survives and differentiates into memory cells, which are long-lived, self-renewing cells that afford increased protection against secondary encounter with the pathogen (1). How differenti-ation into effector and memory cells occurs is still incompletely un-derstood, although the contribution of specific transcription factors (2), metabolic profiles (3–7), epigenetic changes during the course of immune responses (8, 9), and asymmetric cell division (ACD) (10–14) has been explored.

The contact interface between the antigen-presenting cell (APC) and the engaged T cell is termed immunological synapse. By estab-lishing a stable immunological synapse, T cells undergo an extensive cytoskeleton remodeling to build a global polarization axis (15, 16), leading to the asymmetric partitioning of surface molecules, such as the T cell receptor (TCR), CD4/CD8, cytokine receptors, cell fate determinants such as transcription factors, and metabolism such as mTOR (mammalian target of rapamycin) activity (13, 14). The two emerging progenies exhibit different fate potentials (11) and transcrip-tional profiles (10, 17), with the proximal daughter adopting an effector cell fate and the distal daughter being destined to become a memory cell (10, 18). Most studies analyzing ACD have concentrated on naïve murine T cells undergoing their first mitosis after TCR engagement. However, with the exception of memory CD8+ T cells, which were also

reported to undergo ACD (19), it is largely unknown whether other, more terminally differentiated, antigen-experienced T cell subsets, such as effector or exhausted CD8+ T cells, can undergo ACD. Further-more, it also remains to be established whether ACD can be modulated with any functional consequence for T cell fate determination.

In this study, we report that ACD is an ability linked to cell stem-ness, as only naïve and memory CD8+ T cells could divide asymmetri-cally, whereas terminally differentiated effector cells and exhausted cells, which are unable to form a self-renewing reservoir, divided sym-metrically. We identified a strategy to enforce ACD by transient in-hibition of mTOR by rapamycin. Progenies emerging from enforced ACD conditions exhibited increased memory potential because they showed improved survival and expansion upon adoptive transfer, resulting in more efficient protection upon viral challenge. Further-more, transcriptional profiling of progenies derived from symmetric and asymmetric divisions revealed strengthened memory signatures in “memory daughters” emerging from enforced ACD. Functionally, the early memory signature endowed these cells with improved sur-vival and homing to secondary lymphoid organs. Together, these data define ACD as a key mechanism for differentiation and establish-ment of a memory reservoir in murine and human CD8+ T cells.

RESULTSACD is a feature of CD8+ T cells that retain stemnessAs previous reports about ACD in lymphocytes focused on naïve and memory T cells, we investigated whether CD8+ T cells in other differ-entiation stages would also divide asymmetrically. To this end, we adoptively transferred naïve TCR transgenic P14 CD8+ T cells, which spe-cifically recognize the gp33–41 peptide from the lymphocytic chorio-meningitis virus (LCMV) glycoprotein, followed by acute or chronic LCMV infection to generate different CD8+ T cell subsets. Effector cells were isolated 8 days after LCMV WE infection, memory cells were isolated at least 30 days after LCMV WE infection, and exhausted cells

1Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland. 2Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland.*These authors contributed equally to this work.†Corresponding author. Email: [email protected]

Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 2: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

2 of 15

were isolated 30 days after LCMV C13 infection. For analysis of naïve cells, we directly isolated CD8+ T cells from naïve P14 mice splenocytes. After stimulation on -CD3–, -CD28–, and human Fc–ICAM-1 (intercellular adhesion molecule–1)–coated plates for 30 to 36 hours, cells could be identified in different stages of mitosis and analyzed by confocal microscopy. No cell cycle synchronization was used in this work, thus avoiding any perturbation in the cyto-skeleton network having an influence on the generation of asymmetry. We used CD8 cell surface distribution as a readout, based on previous reports that have shown the preferential polarization of this mole-cule toward the proximal daughter (11–14, 18), and confirmed that naïve and memory CD8+ T cells could divide asymmetrically. By contrast, cells isolated from the peak of an acute LCMV infection, composed mostly of short-lived effector cells (SLECs), and exhausted cells, derived from a chronic infection, lacked the ability to generate asymmetry upon mitosis (Fig. 1, A to C). As it has been shown that the integrin ICAM-1 is important for establishment of asymmetry, we also submitted naïve cells to in vitro culture stimulation in the absence of plate-bound human Fc-ICAM-1 and confirmed that this resulted in a marked decrease of asymmetry rates (Fig. 1D). Our experimental observations concerning CD8 distribution in symmetric cell division (SCD) conditions led us to arbitrarily classify a mitosis as asymmetric when one of the daughter cells inherited 50% more CD8 than the other one. To clarify whether the in vitro stimulation using agonistic antibodies was equivalent to antigen presentation by APC, we stimulated P14 CD8+ T cells in a coculture with gp33–41 peptide–loaded dendritic cells. We observed comparable ACD rates under both conditions (fig. S1).

Because we had used bulk populations in our first experiments, we further analyzed whether specific subpopulations within these bulk populations would differ in ACD rates. Specifically, effector P14 cells from 8 days after LCMV WE infection were sorted into SLECs [killer cell lectin-like receptor subfamily G member 1 (KLRG1) interleukin-7 receptor alpha chain (IL-7R−)] or memory precursor effector cells (MPECs; KLRG1− IL-7R+); P14 cells from 30 days after LCMV WE infection were sorted into effector memory cells (EM; CD44+ CD62L−) or central memory cells (CM; CD44+ CD62L+); and P14 cells from 30 days after chronic LCMV C13 infection were sorted into PD-1int (CD44hi) exhausted cells that still retain moderate prolif-erative capacity and are known to be reinvigorated upon programmed cell death protein 1 (PD-1): programmed death ligand 1 (PDL1) blockade or terminally exhausted PD-1hi (CD44lo) cells (20). We ob-served that within the population of effector cells found 8 days after LCMV infection, ACD rates were higher for MPECs in comparison with SLECs. Furthermore, within the memory pool of cells, CM CD8+ T cells divided more asymmetrically than EM CD8+ T cells, whereas for PD-1int or PD-1hi, no asymmetry was observed (Fig. 1, E to G). These results support the concept that ACD correlated with cellular stem-ness, a feature of naïve and memory cells.

Transient mTOR inhibition enhances and restores ACD capabilityAs we observed a correlation between cell stemness and the ability of a CD8+ T cell to undergo ACD, we investigated whether we could modulate asymmetry rates in the different subsets of CD8+ T cells. mTOR inhibition has been reported in T cells to benefit memory formation (21, 22), and the inhibition of the homologous TOR path-way leads to higher asymmetry rates in yeast cells (23). mTOR signals via two different complexes, mTORC1 and mTORC2 (24), containing

distinct scaffold-associated proteins, Raptor and Rictor, which define the downstream targets of each complex. mTORC1 activity is typically measured by the phosphorylation of S6 kinase -1 (P-S6), whereas mTORC2 activity is commonly quantified by phosphorylation of the kinase Akt at serine 473 (S473) (25). We therefore transiently inhib-ited mTOR by treatment with rapamycin (preferential inhibitor of mTORC1) or Akt-kinase inhibitor (which specifically inhibits phos-phorylation of Akt at S473, downstream of mTORC2) (fig. S2).

In activated naïve cells, we observed increased CD8 and T-bet asymmetry in mTOR-inhibited daughter cells, under both transient rapamycin and Akt-kinase inhibitor treatments (Fig. 2, A to C, and fig. S3). mTOR inhibition did not induce ACD in the absence of plate-bound Fc-ICAM-1, suggesting that the polarization initiated by the immune synapse, which is disturbed in the absence of ICAM-1, is necessary to generate the asymmetry that is maintained during mitosis and enhanced by transient mTOR inhibition. Transient inhibition after initial TCR activation was crucial to promote ACD rates, because continuous mTOR inhibition partially prevented mitosis and, within mitotic cells, did not increase ACD rates in comparison with untreated cells (fig. S4A). Similar results to con-tinuous rapamycin treatment were obtained when CD8+ T cells from CD4-cre Raptorfl/fl or CD4-cre Rictorfl/fl mice (lacking mTORC1 and mTORC2 activity in T cells) were stimulated in vitro under ACD conditions (fig. S4B). The transient nature of mTOR inhibition was further confirmed by washing out rapamycin from the cell culture medium 30 to 36 hours after activation and by evaluating P-S6 levels 8 hours later, when we observed reestablishment of its expression with preserved asymmetry (fig. S4C).

After establishing an efficient method of increasing asymmetry in naïve CD8+ T cells, the same approach was applied to the CD8+ T cells in other differentiation stages, using rapamycin as mTOR inhibitor (Fig. 2, D to F). CD8 asymmetry rates were increased in memory pre-cursor cells, EM, and CM, but SLECs continued to divide symmetri-cally. However, more unexpected was the fact that among the exhausted cells, PD-1int CD8+ T cells gained the ability to divide asymmetrically upon transient mTOR inhibition. As TCF-1 was reported to be ex-pressed in a population of memory-like T cells that sustains T cell re-sponses in chronic infections (26), we sorted PD-1int CD8+ T cells into PD-1int TCF-1lo and PD-1int TCF-1hi cells. We observed that TCF-1hi cells can divide asymmetrically and show increased asymmetry rates upon transient rapamycin treatment during stimulation (Fig. 2G). Moreover, similarly to what was observed for naïve CD8+ T cells, higher T-bet asymmetry rates were also observed upon transient mTOR inhibition for MPECs and PD-1int CD8+ T cells (fig. S3). To-gether, these results show that ACD can be modulated in asymmetri-cally dividing cells by short-term mTOR inhibition during mitosis.

Higher ACD rates lead to better memory potential and viral controlHaving established a mechanism to modulate ACD rates, we inves-tigated whether higher asymmetry rates would have any functional consequences. We stimulated naïve P14 cells under asymmetric or symmetric conditions with or without transient rapamycin treatment before transfer into naïve C57BL/6 recipient mice. Recipients were infected with LCMV WE 30 days after transfer (Fig. 3A). We moni-tored the in vivo expansion of the adoptively transferred P14 cells in the blood and spleen of recipient mice, reporting on both survival of the cells in the absence of antigen before challenge and their secondary expansion potential. We consistently observed higher frequencies of

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 3: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

3 of 15

progenies derived from cells that had been stimulated in vitro in the presence of human Fc-ICAM-1 and transiently treated with mTOR inhibitors (Fig. 3, B and C). This increase was partially accompanied by phenotypic changes, with P14 cells submitted to transient mTOR inhibition showing significantly lower frequencies of KLRG1hi IL-7Rlo cells after in vivo recall, and a trend toward higher frequen-cies of KLRG1lo IL-7Rhi cells (Fig. 3D). However, when effector cytokine production was assessed, we observed no differences in

frequencies of interferon- (IFN-) or tumor necrosis factor (TNF)–expressing P14 cells. The higher numbers of total cytokine pro-ducers were a consequence of the higher numbers of P14 cells after in vivo recall (Fig. 3E).

Next, we screened for other drugs that target downstream effec-tors of mTOR (fig. S5A). We found that similar to rapamycin, inhi-bition of hypoxia-inducible factor–1 (HIF-1) enhanced ACD rates (fig S5A). Conversely, FTY720, a sphingosine-1-phosphate

****N

aïve

WE

8 d

piW

E 3

0 dp

iC

13 3

0 dp

iA CD8

DAPI MergeT-betDAPI

β-TubulinDAPI

B C

Naï

veW

E 8

dpi

WE

30

dpi

C13

30

dpi

E FSLECs MPECs EM CM

CD

8D

AP

IM

erge

PD-1hiPD-1int

G

AsymmetricSymmetric

D

(P1

− P

2)/(P

1 +

P2)

(P1

− P2

)/(P1

+ P

2)

(P1

− P2

)/(P1

+ P

2)

AsymmetricSymmetric

SLECs MPECs EM CM PD-1hiPD-1int

** *******

******

*****

–1

1

0

–0.5

0.5

0.2

–0.2

–1

1

0

–0.5

0.5

0.2

–0.2

–1

1

0

–0.5

0.5

0.2

–0.2

Naïve

***

CD8

CD8

CD8

Fig. 1. ACD is a feature of CD8+T cell subtypes that retain stemness. (A) Confocal images from fixed samples of CD8+ T cells 32 to 36 hours after in vitro (re-)activation on (+/-) Fc-ICAM-1–, -CD3–, and -CD28–coated plates. Naïve cells (n = 63) were obtained from wild-type C57BL/6 or P14 mice. Effector (n = 60), memory (n = 61), and exhausted (n = 24) cells were obtained upon sorting of adoptively transferred P14 cells after LCMV (WE or C13 strain) infection. Mitotic cells were identified on the basis of nuclear and -tubulin structures and imaged during late anaphase or cytokinesis, where daughter cells could be distinguished. dpi, days post infection. (B) ACD rates are higher in subsets that retain stemness, namely, naïve and memory cells (30 days after LCMV WE infection). CD8 staining was quantified in both daughter cells, and P1 was arbitrarily set up as the pole (daughter cell) with higher amounts of CD8. Mitosis was classified as asymmetric when the amount of CD8 was 50% higher in one daugh-ter cell in comparison with the other one, defining the threshold of 0.2 (dotted line in the graph). Data are represented as mean ± SEM. (C) Percentage of asymmetrically dividing cells from (B). (D) Stimulation in the absence of Fc-ICAM-1 leads to symmetric mitosis. Naïve CD8+ T cells were activated in -CD3– and -CD28–coated plates in the presence or absence of human Fc-ICAM-1. Data are represented as mean ± SEM. (E) Further subdivision of differentiation subsets revealed that SLECs and PD-1hi/low cells lack the ability to divide asymmetrically. Different cell subsets were obtained upon sorting of adoptively transferred P14 cells 8 days after LCMV WE infection (SLECs, n = 38; MPECs, n = 30), 30 days after LCMV WE infection (EM, n = 23; CM, n = 26) and >30 days after LCMV high-dose C13 infection (PD-1int, n = 47; PD-1hi, n = 32). (F) ACD rates were higher in subsets that retain stemness, MPECs, and MPs. Data are represented as mean ± SEM. (G) Percentage of asymmetrically dividing cells from (F). Statis-tical analysis was performed using the unpaired two-tailed Student’s t test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001 (see also fig. S1).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 4: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

4 of 15

AsymmetricSymmetric

F

–ICAM

–ICAM R

+ICAM

+ICAM R

+ICAM Akt i

AD

AP

Iβ-

Tubu

linM

erge

CD

8–ICAM –ICAM R +ICAM +ICAM R +ICAM Akt i

D

DA

PI

β-Tu

bulin

Mer

geC

D8

SLECs MPECs EM CM PD-1int PD-1hi

**

– + – +R

CD8

E

Asym

met

ric +

ICA

M

Sym

met

ric

Asym

met

ric +

ICA

M R

SLECs

MPECs

EM

CM

PD-1int

PD-1hi

–1.0

–0.5

0.0

0.5

1.0

(P1

− P

2)/(P

1 +

P2)

0.2

–0.2

Day 8 after LCMV WE Day 30 after LCMV WE

– + – +

Day 30 after LCMV C13

– + – +SLECs MPECs EM CM PD-1int PD-1hi

GCD8

DAPIβ-Tubulin

TCF-

1loTC

F-1lo

RTC

F-1hi

TCF-

1hi R

TCF-1hiTCF-1lo– + – +

–1.0

–0.5

0.0

0.5

1.0

(P1

− P

2)/(P

1 +

P2)

0.2

–0.2

Day 30 after LCMV C13 *****

*******

R

B C

(P1

− P

2)/(P

1 +

P2)

***

****

–1

1

0

–0.5

0.5

0.2

–0.2

CD8

+ICAM R

+ICAM Akt

i

+ICAM

–ICAM

–ICAM R

Fig. 2. Transient mTOR inhibition enhances and restores ACD capability. (A) Confocal microscopy images from naïve C57BL/6 or P14 mice CD8+ T cells 32 to 36 hours after stimulation on -CD3– and -CD28–coated plates in the presence or absence of Fc-ICAM-1 [−ICAM, n = 48; −ICAM R, n = 28; +ICAM, n = 90; +ICAM R, n = 80; +ICAM Akt i (Akt-kinase inhibitor), n = 69]. (B) Percentage of asymmetrically dividing cells from (C). (C) ACD rates are higher in cells that were submitted to transient mTOR inhi-bition with both rapamycin or Akt-kinase inhibitor. Data are represented as mean ± SEM. (D) Confocal microscopy images of mitosis from SLECs (n = 12), MPECs (n = 25), EM (n = 13), CM (n = 20), PD-1int (n = 32), and PD-1hi (n = 24) CD8+ T cells after restimulation. (E) Percentage of asymmetrically dividing cells from (F). Frequencies of sym-metrically and asymmetrically dividing cells from conditions with no pharmacological intervention (+ICAM) are depicted, respectively, in white and gray. Frequencies of asymmetrically dividing cells upon transient rapamycin treatment (+ICAM R) are shown in black. (F) ACD is increased upon transient mTOR inhibition in MPECs, EM, and CM CD8+ T cells, and reestablished in PD-1int CD8+ T cells. Asymmetry rates shown in Fig. 1F (white) are depicted on the left side of data from the same subsets obtained upon transient mTOR inhibition (black). Data are represented as mean ± SEM. (G) Confocal microscopy images of mitoses of PD-1int TCF-1lo or TCF-1hi cells submitted or not to transient mTOR inhibition during stimulation (TCF-1lo, n = 16; TCF-1lo R, n = 10; TCF-1hi, n = 15; TCF-1hi R, n = 11). Statistical analysis was performed using the un-paired two-tailed Student’s t test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001 (see also figs. S3 and S4).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 5: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

5 of 15

WEvirus titers

Blood kinetics

LCMV WE200 ffu

A

–30 7 14 21 >28 daysSpleen

0

B

1 20 3Weeks after rechallenge

20

40

60

80

%P

14 c

ells

with

in C

D8

T ce

lls

+ICAM Akt i

+ICAM R

+ICAM

–ICAM ± R

C

% P

14 c

ells

per

spl

een

20

30

40

10

0

% IL

-7R

αloK

LRG

1hiP

14 c

ells

2

4

8

0

6

D

0 103 104 105

010

310

410

5K

LRG

1

IL-7Rα0 103 104 105

010

310

410

5K

LRG

1

32.4

38.1

27.3

42.9

# P1

4 ce

lls p

er s

plee

n

+ICAM

+ICAM R

% IL

-7R

αhiK

LRG

1loP

14 c

ells

20

30

40

10

0

40

60

80

20

0

E*

Activated ± ICAM ±R or Akt i

Naïve P14 cells

*

* *

0.11

% IF

Nγ+

P14

cel

ls%

TN

F+P

14 c

ells

60

80

100

40

0

20

15

20

25

10

0

5

1 ×106

2 ×106

3 ×106

0

# IF

N-γ

+P

14 c

ells

# TN

F+P1

4 ce

lls 6 × 105

8 × 105

0

4 × 105

2 × 105

*

***

IL-7Rα

F

LCMV WE 200 ffu/LCMV C13 2 × 106 ffu

–30 4 12 days0

C13virus titers

WE d4 C13 d12

ffu p

er s

plee

n0 0

108 108

106

104

102

106

104

102 ffupe

r spl

een

** *

Activated± ICAM ±R

Naïve P14 cells

% P

14 c

ells

per

spl

een

# P1

4 ce

lls p

er s

plee

n

0

0.02

0.04

0.06

0.08

5 × 103

0

1 × 104

5 × 104

% P

14 c

ells

per

spl

een

# P1

4 ce

lls p

er s

plee

n

WE C13

0

10

20

30

40

2 × 106

0

4 × 106

6 × 106

* ** **G

0

+ICAM R

+ICAM Akt

i

+ICAM

–ICAM

–ICAM R

*

(×10

6

+ICAM R

+ICAM Akt

i

+ICAM

–ICAM

–ICAM R

*****

0.06 ***0.0534

**

)

Fig. 3. Higher ACD rates lead to better memory potential and viral clearance. (A) Experimental setup. Naïve P14 cells were stimulated under different conditions, harvested after 36 hours, and adoptively transferred into wild-type recipients. Each recipient mouse received 10,000 cells and was infected intravenously with LCMV WE 30 days later. ffu, focus forming units. (B) Frequency of P14 cells within the CD8+ T cell population in the blood of recipients that were infected with LCMV WE 30 days after adoptive transfer. (C) Frequency and numbers of adoptively transferred P14 cells within CD8+ T cells in the spleens of recipient mice. (D) Representative plots of KLRG1 and IL-7R expressing splenic P14 cells stimulated on human Fc-ICAM-1–, -CD3–, and -CD28–coated plates in the presence or absence of rapamycin for 30 to 36 hours before adoptive transfer (left); percentages of KLRG1+ IL-7R− and KLRG1− IL-7R+ P14 cells in the spleens of recipient mice (right). (E) Frequencies and numbers of IFN-– and TNF-producing cells. (F) Top: Experimental setup. P14 naïve CD8+ T cells were stimulated under different experimental conditions for 36 hours and further adoptively transferred to wild-type recipients. Each recipient mouse received 10,000 cells and was infected intravenously with LCMV WE or LCMV C13 30 days after trans-fer. Bottom: Increased memory potential derived from higher ACD rates led to better viral clearance upon LCMV infection. LCMV titers obtained 4 days after infection in spleens of recipient mice that were infected with LCMV WE and 12 days after infection in blood and spleens of recipient mice that were infected with LCMV C13. (G) Fre-quency and numbers of adoptively transferred P14 cells within CD8+ T cells in the spleens of recipient mice. (A to C) Data pooled from five independent experiments. (D) Data pooled from three independent experiments. (E) Representative data from one of two experiments. (F and G) Representative data from one of four experiments. Data are depicted as mean + SEM. Statistical analysis was performed using the unpaired two-tailed Student’s t test or, when data did not pass the normality test, the unpaired Mann-Whitney U test. *P < 0.05; **P < 0.01; ***P < 0.001 (see also figs. S4 to S6).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 6: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

6 of 15

receptor agonist well known for altering lymphocyte trafficking (27, 28) but also a preferential inhibitor of long fatty acid ceramide synthesis (29), decreased ACD rates (fig. S5A). As expected, lower ACD rates resulted in impaired re-expansion potential when activated P14 cells were transferred after in vitro activation to naïve C57BL/6 hosts, fol-lowed by LCMV infection (fig. S5, B and C).

We then tested the protective capacity of progenies from “regular” and enforced ACD after exposure to acute or chronic LCMV infec-tion (Fig. 3F). We found that P14 CD8+ T cells that were transiently exposed to rapamycin upon in vitro stimulation conferred signifi-cantly better protection against infection with both the WE and the C13 strains of LCMV in comparison with untreated progenies (Fig. 3F). The enhanced protection correlated with higher fre-quencies and numbers of P14 cells in the spleens of infected recipient mice (Fig. 3G).

Next, we asked whether an increase of ACD rates by transient rapamycin treatment would also lead to enhanced memory potential in P14 cells in other differentiation states. We therefore stimulated CM P14 cells in vitro with -CD3, -CD28, and human Fc-ICAM-1 in the presence or absence of transient rapamycin treatment, followed by adoptive transfer into naïve wild-type recipients and LCMV re-challenge 30 days later (fig. S6A). Similar to naïve P14 cells, transient rapamycin treatment not only increased ACD rates but also con-ferred enhanced memory potential to CM, reflected by the increased frequencies within CD8+ T cells and total numbers of P14 cells in the blood, spleen, and lungs of recipient mice after acute LCMV infection (fig. S6, B and C). Also in line with what was observed for naïve cells, frequencies of cytokine-producing cells were similar among progenies from all in vitro experimental conditions (fig. S6D). Together, these data provide strong evidence that the enhanced fre-quencies and numbers of P14 cells after recall antigen exposure cor-relate with enforced ability to undergo ACD.

The polarisome is required for ACD modulation by transient mTOR inhibitionTo provide a mechanistic link between (enforced) ACD and mem-ory potential, we sought interventions that would abolish ACD and would allow us to evaluate its functional consequences. Polarisome activity and establishment of polarity depend on protein kinase C (PKC) activity (10, 12, 30). We therefore abrogated polarisome activity by inhibiting PKC with aurothiomalate (AuTM) (31, 32). Naïve CD8+ P14 T cells were activated in vitro with -CD3, -CD28, and human Fc-ICAM-1 in the presence or absence of rapamycin and/or AuTM (Fig. 4A). We observed that inhibiting PKC, rapa-mycin treatment was not able to increase asymmetric distribution of both CD8 and T-bet (Fig. 4, B and C). We then tested whether abrogation of ability to divide asymmetrically due to polarisome disruption would affect memory potential, even in cells that were exposed to transient rapamycin treatment. For that, P14 naïve CD8+ T cells were stimulated under enforced ACD conditions in the presence or absence of AuTM, and their progenies were adop-tively transferred to wild-type recipients, followed by LCMV WE infection after 30 days (Fig. 4D). We observed significantly higher frequencies and numbers of P14 cells in the spleens and lymph nodes of mice that had received progenies from enforced ACD conditions in the absence of AuTM compared with mice that had received progenies from ACD conditions in the presence of AuTM (Fig. 4E). These data provide strong evidence for a causal link be-tween memory potential and the ability to undergo ACD.

Increased ACD and memory potential can be induced in exhausted CD8+ T cells expressing intermediate levels of the co-inhibitory receptor PD-1The robust improvement in memory CD8+ T cell potential in naïve and CM by enforced ACD led us to investigate whether we could also reestablish memory potential in PD-1int cells, because this subset responded to transient rapamycin treatment by gaining, to some extent, the ability to divide asymmetrically. P14 PD-1int cells were stimulated in vitro with -CD3, -CD28, and human Fc-ICAM-1 in the presence or absence of transient rapamycin treatment, and the resulting progenies were transferred into new naïve mice that were rechallenged with LCMV WE 30 days later (Fig. 5A). We found that the reestablishment of ACD resulted in a considerable gain of re-expansion potential upon LCMV infection (Fig. 5, B and C). Of relevance, this re-expansion potential was only seen in cells that were stimulated in the presence of plate-bound Fc-ICAM-1 upon transient mTOR inhibition, also the only condition where PD-1int cells were able to undergo ACD. Besides a drastic numeric increase, these cells also exhibited increased memory markers, characterized by the expression of IL-7R (Fig. 5D), and higher frequencies of effector cytokine (IFN-, TNF)–producing cells (Fig. 5E). Thus, the restoration of effector functions in a previously exhausted T cell population not only corroborates the idea that install-ing ACD leads to an improved memory potential but also opens new perspectives for therapies that could reinvigorate effector responses in chronically infected individuals.

Human naïve and CM CD8+ T cells show higher ACD rates upon transient mTOR inhibitionWe next examined whether human lymphocytes would respond to transient mTOR inhibition similarly to murine cells. Using identical in vitro conditions for stimulation and cell culture as previously de-scribed for CD8+ T cells from mice, human naïve (CD45RA+, CD62L+) (Fig. 6, A to C) and CM (CD45RO+, CD62L+) (Fig. 6, D to F) CD8+ T cells showed increased asymmetry rates upon transient mTOR in-hibition when cultured in the presence of plate-bound Fc-ICAM-1. Increased ACD rates led to better survival when cells were kept in in vitro culture under limiting IL-15 conditions (fig. S7). Thus, tran-sient rapamycin treatment can modulate asymmetry also in human CD8+ T cells, being a potentially useful tool to enhance memory T cell generation in adoptive transfer settings.

ACD generates progenies with strengthened memory signaturesTo experimentally assess whether the better maintenance and/or proliferation potential exhibited by progenies derived from en-forced ACD conditions resulted from higher ACD rates or as a “side effect” of rapamycin treatment, we sorted P14 CD8hi and CD8lo daughter cells after their first mitosis following in vitro stimulation with or without transient mTOR inhibition. Cells were then trans-ferred to naïve C57BL/6 recipients, which were LCMV-infected 30 days later. Quantification of P14 frequencies 1 to 3 weeks after in-fection in the blood of recipients showed that CD8lo cells expanded significantly better than CD8hi cells, irrespective of previous rapa-mycin treatment, confirming that the increased ACD rates imposed by transient rapamycin treatment inferred enhanced memory po-tential to CD8+ T cells (Fig. 7A). These results corroborate previous observations on a better memory potential inherited by CD8lo daughter cells (11, 13).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 7: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

7 of 15

To gain mechanistic insights into the processes that endow CD8+ T cells with increased memory potential upon enforced ACD, we com-pared the transcriptional profiles of CD8lo (potential memory fate) and CD8hi (potential effector fate) first daughter cells derived from prog-enies of naïve CD8+ T cells that were stimulated under SCD (no Fc-ICAM-1) or ACD (in the presence of Fc-ICAM-1) conditions in the presence or absence of transient mTOR inhibition. The distinct gene expression profiles presented by these eight different daughter cell populations reflected their original CD8 expression profiles, classifying them as “effector daughters” (CD8hi) or “memory daughters” (CD8lo), but also whether plate-bound Fc-ICAM and transient rapamycin treat-ment were present or not during stimulation. Within CD8lo and CD8hi cells, gene signatures were shared despite the nature of the stimulation they originated from. However, differences between CD8lo and CD8hi daughters were more pronounced under ACD stimulation conditions, confirming the contribution of this mechanism to generate diversity (Fig. 7B). To address whether distinct memory signatures would be en-riched in CD8lo daughters and particularly within CD8lo daughters emerging after enforced ACD, we quantified the expression of transcrip-tion factors classically associated with effector and memory CD8+ T cell

fates. As expected, CD8lo daughters showed higher expression of Tcf7 and Bcl6, whereas CD8hi cells expressed more Tbx21 and Hif1a (Fig. 7C). The differences between CD8lo and CD8hi cells were stronger among daughter cells emerging from ACD conditions compared with those emerging from SCD conditions, and CD8lo daughter cells derived from enforced ACD showed the most pronounced memory phenotype. Furthermore, expression of genes linked to homing and survival, fea-tures of memory cells, followed a similar trend, with CD8lo cells origi-nating from enforced ACD conditions showing higher expression of Il7r, Sell, Ccr7, and Cxcr3 (Fig. 7D). Sell and Cxcr3, as a number of other genes, were highly influenced by rapamycin treatment, defining a synergistic impact of ACD and mTOR inhibition on the gene signa-ture of CD8lo cells originating from enforced ACD conditions, explain-ing their better memory potential. ACD also led to distinct profiles of cyclin expression. CD8lo cells expressed considerably lower levels of Ccnd2, Ccnd3, and Cdc25c compared with CD8hi cells (fig. S8). This gene expression profile was supported by functional data, as the proliferation rates observed for progenies of naïve CD8+ T cells com-ing from enforced ACD conditions were slower than the ones ob-served in standard ACD conditions or SCD conditions (fig. S8).

P-ERK

A

0 103 104 105

AuTMControl

B DAPIβ-Tubulin MergeCD8T-bet

+R+A

uTM+A

uTM R

C

(P1

− P

2)/(P

1 +

P2)

–1

1

0

–0.5

0.5

0.2

–0.2

CD8

+R

+AuT

M

+AuT

M R

(P1

− P

2)/(P

1 +

P2)

–1

1

0

–0.5

0.5

0.2

–0.2

T-bet

+R

+AuT

M

+AuT

M R

D

LCMV WE200 ffu

–30 10 days

Organs

0

+ICAM ±R ±AuTM activated naïve P14 cells

E

+R

+AuT

M

+AuT

M R +R

+AuT

M

+AuT

M R +R

+AuT

M

+AuT

M R +R

+AuT

M

+AuT

M R

% P

14 c

ells

per

spl

een

# P

14 c

ells

per

spl

een

0

2

4

6

0

5

10

15

20

25

0

5 × 10

1.5 × 10

1 × 10

2

3

3

% P

14 c

ells

per

LN

# P

14 c

ells

per

spl

een

0

2 × 10

1 × 105

4

4 × 104

6 × 104

8 × 104

********

***

***

0.11

**

**

Mod

al

Fig. 4. The polarisome is required for ACD modulation by transient mTOR inhibition. (A) Inhibition of phosphorylated extracellular signal–regulated kinase (P-ERK) was used as a readout for AuTM efficiency on PKC inhibition. (B) Confocal microscopy images from stimulated naïve CD8+ T cells obtained from P14 mice (+ICAM R, n = 52; +ICAM AuTM, n = 33; +ICAM AuTM R, n = 32). (C) CD8 and T-bet asymmetry upon transient PKC inhibition. Data are represented as mean ± SEM. (D) Experimental setup. P14 naïve CD8+ T cells were stimulated under different experimental conditions for 36 hours and adoptively transferred to wild-type recipients. Each mouse received 10,000 cells and was infected intravenously with LCMV WE 30 days after transfer. (E) Frequencies and numbers of adoptively transferred P14 cells within CD8+ T cells in the spleens and lymph nodes (LNs) of recipient mice 10 days after LCMV infection. Data are depicted as mean + SEM. (A to C) Data pooled from two independent experi-ments. (D and E) Representative data from one of two experiments. Statistical analysis was performed using the unpaired two-tailed Student’s t test or, when data did not pass the normality test, the unpaired Mann-Whitney U test. *P < 0.05; **P < 0.01; ****P < 0.0001.

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 8: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

8 of 15

Distinct gene signatures of fully formed effector and memory CD8+ T cells have been extensively described. Recently, defined expression clusters being up- or down-regulated in effector cells (KLRG1+ IL-7R−, 8 days after LCMV infection) in comparison with memory cells (CD44+CD62L+, 60 days after LCMV infection)

were reported (33). We compared the transcriptional profile of first CD8lo and CD8hi daughter cells emerging from SCD, standard ACD, and enforced ACD stimulation conditions with the expres-sion profiles of fully differentiated effector and memory cells. CD8hi daughter cells presented a transcriptional profile more similar to

A B

LCMV WE200 ffu

–30 7 14 21 >28 days

Blood kinetics Organs

0

Activated± ICAM ±R

PD-1int P14 cells

1 20 3Weeks after rechallenge

+ICAM R

+ICAM –ICAM ±R %

P14

cel

ls w

ithin

CD

8 T

cells 6

4

2

0

% P

14 c

ells

per

spl

een

# P1

4 ce

lls p

er s

plee

n

4 × 105

0

1 × 106

0

1

2

3

8 × 105

6 × 105

2 × 10 5

% IF

N-γ

+P

14 c

ells

40

0

20

60

80

% T

NF+

P14

cells 40

0

10

50

**

C

E

# IF

N-γ

+P

14 c

ells

# TN

F+P1

4 ce

lls

5 × 104

1 × 105

2 × 105

0

1.5 × 105

5 × 104

1 × 105

0

1.5 × 105

****

******

ns

*****

ns

% P

14 c

ells

per

lung

# P1

4 ce

lls p

er lu

ng

2 × 105

00

2

4

6 4 × 105

3 × 105

1 × 105

****

ns

****

ns

% IL

-7R

αloK

LRG

1hiP1

4 ce

lls

40

60

20

0

% IL

-7R

αhiK

LRG

1loP1

4 ce

lls

10

5

0

15

20

25

ns

*

20

30

0.10

2.5 × 105

***

***

D

0 103 104 105

010

310

410

5K

LRG

1

0 103 104 105

010

310

410

5K

LRG

1

41.5

24.6

15.8

41.6 +ICAM

+ICAM R

IL-7Rα

IL-7Rα

Fig. 5. Exhausted CD8+T cells expressing intermediate levels of the co-inhibitory receptor PD-1 show recovered memory potential when ACD capacity is rees-tablished. (A) Experimental setup. P14 exhausted CD8+ T cells (PD-1int CD44hi cells isolated from spleens of recipient mice >30 days after LCMV C13 infection) were stimulated under different experimental conditions for 36 hours and further adoptively transferred to wild-type recipients. Each mouse received 10,000 cells and was infected with LCMV WE 30 days after transfer. (B) Frequency of P14 cells within the CD8+ T cell population in the blood. (C) Frequency and numbers of adoptively trans-ferred P14 cells within CD8+ T cells in the spleens and lungs. (D) Representative plots of KLRG1 and IL-7R expressing splenic P14 cells stimulated on human Fc-ICAM-1–, -CD3–, and -CD28–coated plates in the presence or absence of rapamycin for 30 to 36 hours before adoptive transfer (left); percentages of KLRG1+ IL-7R− and KLRG1− IL-7R+ P14 cells in the spleens of recipient mice (right). (E) Frequencies and numbers of IFN-– and TNF-producing cells. Data from two pooled experiments. Data are depicted as mean + SEM. Statistical analysis was performed using the unpaired two-tailed Student’s t test or, when data did not pass the normality test, the unpaired Mann-Whitney U test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 9: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

9 of 15

that of effector CD8+ T cells than their CD8lo counterparts (fig. S9, left). Among the CD8lo cells, the ones derived from ACD exhibited gene signatures closer to memory cells, especially the ones derived from enforced ACD conditions (fig. S9, right). It needs to be em-phasized, however, that the differentially expressed genes defining “effector” and “memory” clusters were established by a comparison between CD8+ T cells that were much further in their differentia-tion than first daughter cells (33). As such, it is remarkable that already first daughter cells tend to show transcriptional similarities to their future to be differentiation states. The only available tran-scriptional profiling of first daughter cells previously reported used single-cell RNA sequencing data (17), which make direct compari-sons technically challenging owing to significant differences in se-

quencing depth. To obtain a better insight into an early (potential) memory gene signature among the CD8lo daughter cells, we se-lected 50 genes linked to T cell differentiation that were differen-tially expressed both in our dataset (first daughter cells) and in the one from He et al. (33) (fully formed effector and memory cells). Among the CD8hi cells, the ones deriving from standard ACD con-ditions had a gene signature more similar to effector cells, whereas enforced ACD gave rise to CD8lo progenies with a stronger memory signature (Fig. 7E). Moreover, the differences in gene expression between CD8lo and CD8hi cells were generally higher between daughters deriving from ACD conditions (Fig. 7F), confirming a role for ACD in the generation of diversity and cells with enhanced memory potential.

DA

PI

β-Tu

bulin

Mer

geC

D8

–ICAM –ICAM R +ICAM +ICAM RB C

–IC

AM

–IC

AM

R

+IC

AM

+IC

AM

R

E FD

DA

PI

β-Tu

bulin

Mer

geC

D8

–ICAM –ICAM R +ICAM +ICAM R

–IC

AM

–IC

AM

R

+IC

AM

+IC

AM

R

A

AsymmetricSymmetric

AsymmetricSymmetric(P

1 −

P2)/(

P1 +

P2)

(P1

− P2

)/(P1

+ P

2)

***

*

–1

1

0

–0.5

0.5

0.2

–0.2

–1

1

0

0.5

0.2

–0.5

–0.2

Naï

ve C

D8+

T ce

llsM

emor

y C

D8+ T

cel

ls

CD

8C

D8

ICAMR

– +––

+– + +

ICAMR

– +––

+– + +

Fig. 6. Human naïve and CM CD8+T cells show higher ACD rates upon transient mTOR inhibition. (A) Confocal images from fixed samples of human naïve CD8+ T cells 32 to 36 hours after in vitro stimulation. Transient mTOR inhibition (12 hours after activation until fixation) was achieved by treatment with 20 nM rapamycin. Stimulation was done on -CD3– and -CD28–coated plates in the presence or absence of Fc-ICAM-1 (−ICAM, n = 11; −ICAM R, n = 10; +ICAM, n = 37; +ICAM R, n = 42). (B) ACD rates in the presence or absence of transient mTOR inhibition with rapamycin. Data are represented as mean ± SEM. (C) Percentage of asymmetrically dividing cells from (B). (D) Confocal images from fixed samples of human CM CD8+ T cells 32 to 36 hours after in vitro stimulation. Transient mTOR inhibition (12 hours after acti-vation until fixation) was obtained upon treatment with 20 nM rapamycin. Stimulation was done on -CD3– and -CD28–coated plates in the presence or absence of Fc-ICAM-1 (−ICAM, n = 7; −ICAM R, n = 7; +ICAM, n = 15; +ICAM R, n = 19). (E) ACD rates in cells that were submitted to transient mTOR with rapamycin. Data are repre-sented as mean ± SEM. (F) Percentage of asymmetrically dividing cells from (E). Pooled data from peripheral blood mononuclear cells of four different healthy donors. Statistical analysis was performed using the unpaired two-tailed Student’s t test. *P < 0.05; **P < 0.01 (see also fig. S5).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 10: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

10 of 15

−0.6 −0.4 −0.2 0.0 0.2 0.4 0.6

−0.6

−0.4

−0.2

0.0

0.2

0.4

0.6

PC1

PC

2

−3 −2 −1 0 1 2 3

−3−2

−10

12

3

Tbx21

Irf4

LifIl2ra

Il2rb

Il12rb2

Itgb2

Ezh2

Hif1a

Rheb

Hk2

Gzmb

Ccnd2

Ccnd3

Cdc25c

Casp7

Idh3aPdcd1

Ctla4

Slc7a5

Sell

Cxcr3Ccr2Ccr7 Ccl5

Il7r

Cd7

Tcf7

Bcl6

Id2

Foxo1

Eomes

Itgb7Itgal

Itgax

Xcl1

S1pr1

Mcl1

Prss12

Itga4

Smad7Slc30a1

Klf2

Lef1

Nr4a1Ccl9

Cxcl10Cd5

Il23a

Adam8

E

M

1

23

4

5

6

7

8

9.5

CD8hi

StimulatedUnstimulated

CD8hi R

15

20

10

0

5

1 2 3

*

0.055ns

Weeks after rechallenge

0 103 104 105CTY

Mod

al

1st daughters

0 104 105CD8

CD8lo CD8lo R

hilo

%P

14 c

ells

with

in C

D8

T c

ells

Blood

A B

ICAM – + – + – + – +– –R – –+ + + +

CD8hiCD8lo

C

Mod

al

8.0

8.5

9.0

10.0

Log

CPM

lo++

lo lo hihihihi loCD8– – + +––ICAM

R +–– + – ++–

Tcf7

Bcl6

1.5

Log

CPM

2.5

3.5

4.5

1 2 3 4 5 6 7 8 1 2 3 4 5 6 7 8

Tbx21

6.0

8.0

Log

CPM

1 2 3 4 5 6 7 8

6.5

7.0

7.5

Hif1a

1 2 3 4 5 6 7 8

7.0Log

CP

M

7.5

8.0

8.5

6.5

D

Il7r

4.0

Log

CP

M

5.0

6.0

7.0

1 2 3 4 5 6 7 8 1 2 3 4 5 6 7 8

Sell

Log

CP

M

7.0

8.0

9.0

10.0

1 2 3 4 5 6 7 8

Ccr7

Log

CPM

9.0

9.5

10.0

10.5

11.0

1 2 3 4 5 6 7 8

Cxcr3

Log

CP

M

9.0

9.5

10.0

10.5

11.0

E F Il23aNr4a1Xcl1Cxcl10Adam8Id2EomesCcr2Slc30a1Cd7Smad7Klf2Ccr7Mcl1Il7rS1pr1ItgaxCd5Bcl6Ccl9Prss12Itga4Cxcr3Tcf7SellItgb7Ccl5Lef1Pdcd1Foxo1ItgalCasp7Ctla4Ccnd2Il2raHif1aCdc25cIl12rb2Ezh2Itgb2Il2rbLifCcnd3Irf4GzmbIdh3aRhebTbx21Hk2Slc7a5

1

2

3

4

8

Effector CD8+ T cells

Memory CD8+ T cells

–ICAM CD8lo

–ICAM CD8hi

–ICAM R CD8lo

–ICAM R CD8hi

+ICAM CD8lo

+ICAM CD8hi

+ICAM R CD8lo

+ICAM R CD8hi

lo++

lo lo hihihihi loCD8– – + +––ICAM

R +–– + – ++–

M

E

6

7

5

5.52

–2

0

2

–2

0

*+

Fig. 7. ACD generates progenies with strengthened memory signatures. (A) Naïve P14 cells were stimulated on human Fc-ICAM-1–, -CD3–, and -CD28–coated plates in the presence or absence of rapamycin (initiated 12 hours after stimulation) for 30 to 36 hours. Cell trace yellow (CTY) dilution was used to identify first daughter cells, from which CD8lo and CD8hi cells were sorted and transferred to recipient mice. Thirty days after adoptive transfer, recipients were infected with LCMV WE. Frequencies of P14 cells in the blood of mice (bottom); CD8 expression in first daughter cells that were submitted or not to rapamycin treatment before adoptive transfer (top). Represent-ative data from one of two experiments. Data are depicted as mean ± SEM. Statistical analysis was performed using unpaired two-tailed Student’s t test or, when data did not pass the normality test, the unpaired Mann-Whitney U test. ns, not significant. (B) Differential expression of genes among CD8lo and CD8hi first daughter cells (top 100 differ-entially expressed genes based on the lowest P value) presented as a heatmap. (C) Expression of selected transcription factors previously associated with CD8+ T cell differ-entiation into effector or memory cells. (D) Expression of selected genes associated with survival and homing to secondary lymphoid organs, features of long-lived memory cells. (E) Principal components analysis of 50 selected genes previously associated with CD8+ T cell differentiation. Green, effector gene signatures; blue, memory gene signatures. (F) Differential expression of the 50 selected genes among CD8lo and CD8hi first daughter cells derived from SCD, standard ACD, and enforced ACD.

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 11: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

11 of 15

Enforced ACD generates daughter cells with improved homing featuresThe transcriptional profiling of CD8lo and CD8hi daughters origi-nating from distinct stimulation conditions highlighted the increased expression of genes associated with survival and homing to second-ary lymphoid organs in CD8lo cells, particularly after enforced ACD.In vitro, progenies of transiently mTOR-inhibited cells survived better under limiting IL-15 conditions (fig. S7). To functionally cor-roborate whether this would also reflect in improved T cell homing and access to survival niches in vivo, we isolated CD8lo and CD8hi daughter cells from SCD, standard ACD, and enforced ACD stimu-lation conditions, and equal numbers of each type of cells were transferred into naïve hosts. After 6 to 7 hours, we harvested sec-ondary lymphoid organs and peripheral organs from the recipient animals (Fig. 8A). We consistently observed significantly higher fre-quencies and numbers of CD8lo cells in secondary lymphoid organs in comparison with their CD8hi counterparts (Fig. 8B). In the periph-eral organs analyzed (lungs and liver), this was also observed but to a lower extent. CD8lo cells from enforced ACD conditions had the best performance concerning homing to secondary lymphoid organs. To confirm whether the homing would also reflect in access to sur-vival niches, we analyzed sections of spleens from the recipient mice. CD8hi cells could not be found in most of the spleen sections analyzed, and when present, they were mostly localized in the red pulp. CD8lo cells were found in the white pulp, localized mostly in the T cell zone, and were there in significantly higher numbers when originating from enforced ACD (Fig. 8C). To experimentally ad-dress whether better homing to secondary lymphoid organs also reflected in improved maintenance of donor cells, we transferred equal numbers of P14 cell progenies originating from symmetric, asym-metric, or enforced ACD stimulation conditions into naïve wild-type recipients with no further viral challenge. After 4 weeks, we observed significantly higher frequencies of enforced ACD progenies in the spleen and lymph nodes of recipient mice (Fig. 8D). Our results strongly suggest that ACD contributes to the rise of CD8lo cells en-dowed with homing potential to T cell zones of secondary lymphoid organs, a feature that is improved upon enforced ACD and that con-tributes to their survival and the observed increased re- expansion upon antigen reencounter.

DISCUSSIONCD8+ T lymphocytes are an integral part of the adaptive immune system and are equipped with highly variable TCRs, which convey antigen specificity. There is already an immense repertoire of dif-ferent naïve CD8+ T cells before the antigen encounter, but for an immune response to be effective, both clonal expansion and differ-entiation must take place (1). The observation that an individual naïve cell can generate different progenies, in a diversification degree comparable to polyclonal T cells responses, supports the idea that fate is not predetermined (34–36). Furthermore, previous studies have established transcriptional profiles (2, 37), epigenetic control of gene expression (8, 9), usage of preferential metabolic pathways (7), and ACD (38–40) as mechanisms that contribute to differentiation. Generation of diversity through ACD is a conserved mechanism of special relevance for development and cellular adaptation, being reported not only in different metazoan cell types besides lympho-cytes but also in a wide range of other organisms. Asymmetry relies on the establishment of a polarization axis, which is attained in CD8+

T lymphocytes through the interaction with APCs in the immunological synapse. The orientation of the polarity axis and the composition of the two opposite poles are guided by polarity proteins, and asym-metric distribution is built in different layers after T cell activation, leading to the rise of two progenies that differ in phenotype, transcrip-tion factors composition, metabolic status, and transcriptional profile (10, 11, 13, 14, 18) and, as a result, inherit different potential fates. These data are supported both by microscopy observations and by transcriptional profiling of single cells during immune responses, where an early bifurcation of gene expression is detected already after one cycle of mitosis (17).

ACD strongly relies on the formation of a stable immunological synapse. Both absence of ICAM-1 and presentation of low-affinity peptides by APCs do not induce ACD (11, 18), and we have made similar observations when stimulating CD8+ T cells in the absence of Fc-ICAM-1. Thus far, most studies investigating ACD in T lym-phocytes were performed with naïve cells, with one exception re-porting memory cells to undergo ACD (19). Here, we unraveled a correlation between ACD and memory potential. SLECs and termi-nally PD-1hi exhausted CD8+ T cells were unable to divide asym-metrically, whereas cells that retain stemness, i.e., the potential to differentiate into subsets with a different fate, exhibited ACD. Con-sidering the regulatory role of mTOR on CD8+ T cell differentiation and the beneficial impact of mTOR inhibition for memory formation (21, 22), we hypothesized that mTOR inhibition could potentially enhance or reestablish the ability of CD8+ T cells to divide asym-metrically. This hypothesis was also based on studies in yeast, where TOR inhibition led to higher ACD rates (23). We found that tran-sient mTOR inhibition through rapamycin or Akt-kinase inhibitor treatment resulted in higher ACD rates in naïve and memory cells. In addition, it installed the ability to undergo ACD in PD-1int exhausted cells. It is important to highlight that the modulation of ACD was restricted to transient mTOR inhibition after TCR en-gagement. When CD8+ T cells from CD4-cre Raptorfl/fl or CD4-cre Rictorfl/fl mice, constitutively lacking mTORC1 and mTORC2 activity, were stimulated in vitro under ACD conditions, no increase in ACD rates was observed. This observation corroborates previous find-ings, where CD8+ T cells from T-Rheb−/− or T-Tsc2−/− mice presenting constitutively low and high amounts of mTOR activity, respectively, showed similar CD8 asymmetry to wild-type cells (13). Being a key regulator of T cell metabolism (24, 41, 42), it is reasonable to hypothesize that mTOR inhibition has multiple downstream con-sequences. However, we could prove that changes in asymmetry are detached from any permanent change in metabolism because mTOR activity was reestablished few hours after removal of drug treatment.

A further link between (enforced) ACD and the ability to generate progenies with (enhanced) memory potential was obtained by selec-tive polarisome disruption during the first mitosis, as CD8+ T cells stimulated under conditions that normally support ACD and installa-tion of memory potential would fail to do so when PKC, a compo-nent of the polarisome, was inhibited. The polarisome complex is key for the establishment of asymmetry, and ablation or silencing of PKC leads to a general defect in establishing polarity, resulting in defective T cell memory (10, 30). Thus, our results suggest that transient mTOR inhibition is not able to create polarity but benefits the establishment and maintenance of asymmetry in polarizing conditions.

By transiently inhibiting mTOR activity and, consequently, in-creasing ACD rates, adoptively transferred naïve, memory, and PD-1int

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 12: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

12 of 15

exhausted P14 CD8+ T cell progenies showed increased expansion upon LCMV challenge compared with nontreated cells, supporting the idea that asymmetry favors the rise of CD8lo cells that can survive

in the absence of antigen and thereafter respond to antigen rechal-lenge. The transcriptional profiling of CD8lo and CD8hi populations originating from distinct stimulation conditions (SCD, ACD, and

A

6 to 7 hours

CD8lo or CD8hi daughtersfrom ± ICAM ±R

activated naïve P14 cells

homing assessment

2nd lymphoid organsperipheral organs

B

% P

14 c

ells

per

lung

0

0.5

1.0

1.5

% P

14 c

ells

per

live

r

0

0.4

0.5

0.3

0.2

0.1

C

−ICAM +ICAM RCD8hi

CD169 CTY B220 CD8

# P

14 c

ells

per

T c

ell z

one

0

1

2

3

4

% P

14 c

ells

per

spl

een

0

0.05

0.10

0.15

0.20 ***

***

% P

14 c

ells

per

LN

0

0.005

0.010

0.015 *****

****

****

CD8hiCD8lo CD8loCD8

ICAMR

lo lo hi hi− +− +− +− +

CD8ICAM

R

lo lo hi hi− +−

−+

− + +− +−

−+

− + +

hi hilo lo

D± ICAM ±R

activatednaïve P14 cells

Survival assessment

>4 w

eeks

E

% P

14 c

ells

per

spl

een

0

0.05

0.10

0.15

0

2 × 10

6 × 10

8 × 10

# P

14 c

ells

/spl

een

4 × 10

1 × 10

3

3

3

3

4

% P

14 c

ells

per

LN

0

0.05

0.10

0.15

0

5 × 10

1.5 × 10

# P

14 c

ells

per

LN

1 × 10

2

3

3

ICAMR

− +−−

+− + +

− +−−

+− + +

− +−−

+− + +

− +−−

+− + +

**************** **

************

***********

Fig. 8. Enforced ACD generates daughter cells with improved homing features to T cell zones of secondary lymphoid organs. (A) Experimental setup. Naïve P14 cells were stimulated for 30 to 36 hours on -CD3– and -CD28–coated plates, in the presence or absence of plate-bound Fc-ICAM-1 and/or transient mTOR inhibition by rapamycin (starting 12 hours after activation). CTY dilution was used to identify first daughter cells, from which CD8lo and CD8hi cells were sorted and transferred to recip-ient mice. Each recipient mouse received 1 × 105 to 3 × 105 P14 cells. Organs were harvested 6 to 7 hours after transfer. (B) Frequencies of P14 cells within the CD8+ T cell population in spleen, inguinal lymph nodes, lungs, and liver of recipient mice. (C) Right: Confocal images from 10-m splenic sections. Tissues were stained for the local-ization of metallophilic macrophages (CD169), B cells (B220), and CD8+ T cells (CD8). P14 cells could be identified by their preserved CTY staining. Left: Quantification of P14 cells present in the imaged sections of T cell zones from different recipients (−ICAM CD8lo, n = 35; +ICAM R CD8lo, n = 50; −ICAM CD8hi, n = 30; +ICAM R CD8hi, n = 25). (D) Naïve P14 cells were stimulated under different conditions and harvested after 36 hours, and progenies were adoptively transferred into wild-type naïve recipients (1 × 105 cells per mouse). (E) Frequencies and numbers of transferred cells were assessed 4 weeks later in spleen and lymph nodes of recipients. (A and B) Pooled data from five independent experiments. (C) Representative data from one of two experiments. (D and E) Representative data from one of three experiments. Data are depicted as mean + SEM. Statistical analysis was performed using the unpaired two-tailed Student’s t test or, when data did not pass the normality test, the unpaired Mann-Whitney U test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 13: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

13 of 15

enforced ACD) revealed the influence of both ICAM-1 and rapa-mycin as variables that can affect gene signatures. The role of ICAM-1 engagement in providing a stable immunological synapse and pro-longed APC–T cell interactions and the consequences of mTOR inhibition as a tool to improve homing of CD8+ T cell to survival niches and benefit memory differentiation have been previously re-ported (21, 43–46). Our data support the notion that ICAM-1 and mTOR inhibition potentially contribute to alterations in T cell differ-entiation through ACD modulation, as we observed a synergistic effect of these two components on T cell fate determination. Enforced ACD generated CD8lo daughters presenting higher expression of genes linked to memory differentiation. mTOR inhibition has been previously reported as a tool to improve homing of CD8+ T cell to survival niches (43, 44). Here, we could extend this knowledge and determine the role of first CD8lo daughter cells in this process, with progenies of enforced ACD conditions featuring best survival rates and most efficient homing to T cell zones in secondary lymphoid organs. Mechanistically, we suggest that ACD gives rise to more diverse progenies compared with SCD, which could be confirmed by the more distinct gene expression profile differences between CD8lo and CD8hi daughter cells emerging from stimulation condi-tions that benefit ACD. Enforcing ACD by transient rapamycin inhibition imprinted CD8lo progenies with an earlier and stronger memory signature, increasing their access to survival niches and their memory potential. As we could also show that human CD8+ T cells increase ACD rates upon transient rapamycin treatment, these results open new perspectives in the context of vaccinations and possibly adoptive transfer therapies to resolve tumor and chronic infections. Even preterminally exhausted PD-1int CD8+ T cells ex-hibited increased ACD rates and memory potential when transiently treated with rapamycin during reactivation, resulting in recovered expansion potential and effector functions. These results open new opportunities of enforcing ACD as a means to improve memory potential of exhausted CD8+ T cells, with relevance for “rejuvenation” of exhausted CD8+ T cells in the context of chronic infections and cancer.

We acknowledge that despite the potential opportunities of ACD modulation as a tool for boosting memory responses or re-juvenating exhausted CD8+ T cells, the specific mechanisms that drive increased polarization upon transient mTOR inhibition re-main to be unraveled. In this respect, there is a definitive need to further unravel the molecular details that link mTOR inactivation and increased cell polarity via PKC, potentially allowing the dis-covery of more specific ways of promoting ACD without altering other cellular pathways. This quest might not be trivial as mTOR complex activity orchestrates many downstream pathways that affect various aspects of cellular physiology, including cellular metabolism, lipid biosynthesis, cell migration, survival and differ-entiation, proliferation, and protein translation. However, our ob-servation that transient mTOR inhibition during mitosis of T cells retaining stem cell–like potential promotes memory potential, which is linked to increased ACD, provides new opportunities to endow this functional trait to T cells in the context of vaccinations and T cell–based immunotherapies.

MATERIALS AND METHODSA detailed description of materials and methods is provided in the Supplementary Materials.

Study designThis study aimed to evaluate the impact of the differentiation status of a CD8+ T cell on its ability to divide asymmetrically and to deter-mine the potential of ACD modulation as a mechanism to increase or install memory potential on T cells. For that purpose, we stan-dardized an experimental protocol to quantify polarization of dif-ferent molecules upon CD8+ T cell mitosis. We used the LCMV infection model as a tool to generate diversity and isolate different T cell subsets. We implemented adoptive transfers of CD8+ T cell progenies, following first mitosis after (re-)activation, as a readout to measure memory potential imprinted by ACD. All experiments were performed at least twice. Every group consisted of at least two mice. No outliers were excluded from the data analysis, and no ran-domization or blinding was used.

CD8+ T cell in vitro stimulation and adoptive transferCD8+ T cells were cultured in T cell medium (RPMI-1640,BioConcept), 2 mM l-glutamine (BioConcept), 2% penicillin-streptomycin (Sigma- Aldrich), 10% fetal bovine serum (OMNILAB), 25 mM Hepes (Gibco, Life Technologies; Zug, Switzerland), 1× non- essential amino acids (Sigma-Aldrich), 50 M -mercaptoethanol (Gibco), and 1 mM so-dium pyruvate (Gibco) supplemented with self-made human IL-2 and stimulated on -CD3 (5 g/ml) (145-2C11, BioLegend)–coated and -CD28 (5 g/ml) (37.51, BioLegend)–coated plates in the presence or absence of plate-bound human Fc-ICAM-1 (50 g/ml) (R&D Systems, Bio-Techne AG; Zug, Switzerland) for 30 to 36 hours before confocal microscopy analysis or adoptive transfer experi-ments. mTOR modulation was conducted by adding 20 nM rapamycin (Santa Cruz Biotechnology, Labforce AG; Muttenz, Switzerland), 1 to 2 M Akt-kinase inhibitor (Sigma-Aldrich), 1 M c-Myc in-hibitor (10058-F4, Sigma-Aldrich), 1 M HIF-1 inhibitor (FM19G11, Sigma-Aldrich), 1 M FTY720 (Sigma-Aldrich), or 10 M AuTM (Sigma-Aldrich) 12 hours after stimulation. For adoptive transfer, 1 × 104 purified CD45.1 P14 cells (stimulated in vitro or not) were intravenously injected into naïve C57BL/6 CD45.2 recipient mice.

Immunofluorescence staining and confocal microscopyCD8+ T cell lymphocytes previously (re-)activated in vitro were washed in phosphate-buffered saline (PBS) and added on poly-l- lysine (Sigma-Aldrich)–treated coverslips, followed by incubation for 30 min at 37°C. Cells were then fixed with 2% paraformalde-hyde (Sigma-Aldrich) for 10 min, permeabilized with 0.1% Triton X-100 (Sigma-Aldrich) for 10 min, and blocked in PBS containing 2% bovine serum albumin (GE Healthcare) and 0.01% Tween 20 (National Diagnostics) for 1 hour at room temperature. The following anti-bodies were used to perform immunofluorescence staining in murine cells: mouse --tubulin (Sigma-Aldrich), -mouse immuno globulin G Alexa Fluor 488 (Abcam, Lucerna-Chem AG; Luzern, Switzerland), -CD8 APC (53-6.7, BioLegend), -T-bet phycoerythrin (PE) (4B10, BioLegend), and -phospho-S6 PE (D57.2.2E, Cell Signaling Tech-nology). For human cells, we performed tubulin staining with the same antibodies, but -CD8 PE (SK1, BD Biosciences) was used. Mitotic cells (late anaphase to cytokinesis) were identified by nu-clear morphology, the presence of two microtubule organizing centers, and a clear tubulin bridge between two daughter cells. Asym-metry rates were calculated on the basis of CD8 quantification (vol-ume and fluorescence intensity) in each hemisphere. P1 was defined as the pole containing higher amounts of CD8, and P2 was defined

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 14: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

14 of 15

as the opposite pole. Mitoses were considered asymmetric when CD8 enrichment was 1.5-fold greater in one daughter cell in com-parison with the other [(P1 − P2)/(P1 + P2) > 0.2]. For splenic tissue sections, 10-m-thin sections were prepared from frozen spleens embedded in optimal cutting temperature compound (Sakura). Samples were fixed in 2% paraformaldehyde for 1 hour at room tem-perature and blocked in PBS containing 1% of rat serum for 1 hour at room temperature. Staining was performed with the following antibodies (diluted 1:100 in PBS with 0.1% rat serum) for 1 hour at room temperature in the dark: -CD8 BV421 53-6.7, -CD169 Alexa Fluor 647 3D6.112, and -B220 fluorescein isothiocyanate RA3-6B2 (BioLegend). 4′,6- Diamidino-2-phenylindole (Sigma-Aldrich) was used to detect DNA. Mowiol (Calbiochem, Merck- Millipore; Schaffhausen, Germany) was used as mounting medium. Twenty to thirty Z-stacks were acquired using a Visitron Confocal System (inverse confocal microscope, Visitron Systems GmbH) with 10× or 100× magnification (10× objective, type: Plan-Neofluar, aperture: 0.3; immersion: air, contrast: Ph1; 100× objective, type: Plan-Neofluar, aperture: 1.3; immersion: air, contrast: Ph3) coupled to Evolve 512 EMCCD cameras (Photometrics). Data were analyzed using Volocity software (version 6.3., PerkinElmer).

Statistical analysisTo test whether data point values were distributed in a Gaussian dis-tribution, we performed D’Agostino-Pearson normality test. For statistical analysis, we performed two-tailed Student’s t test or non-parametrical analysis Mann-Whitney U tests using GraphPad Prism Software (version 7.0). Statistical significance was determined with *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. For animal experiments, sample size varied from two to seven mice per group for each individual experiment.

SUPPLEMENTARY MATERIALSimmunology.sciencemag.org/cgi/content/full/4/34/eaav1730/DC1Material and MethodsFig. S1. Stimulation of P14 CD8+ T cells on -CD3–, -CD28–, and Fc-ICAM-1–coated plates or in a coculture with dendritic cells leads to comparable ACD rates.Fig. S2. Transient mTOR inhibition was obtained by treatment with rapamycin or Akt-kinase inhibitor.Fig. S3. Transient mTOR inhibition modulates T-bet asymmetry.Fig. S4. Transient but not continuous mTOR inhibition is a requirement for ACD modulation in CD8+ T cells.Fig. S5. Decreased ACD rates in P14 CD8+ T cells submitted to FTY720 treatment led to reduced proliferation upon LCMV challenge.Fig. S6. CM CD8+ T cells produce progenies with enhanced memory potential after enforcing ACD.Fig. S7. Murine and human rapamycin-treated naïve CD8+ T cells produce progenies that can survive better in the presence of limited amounts of IL-15.Fig. S8. CD8lo and CD8hi daughter cells inherit distinct cyclin expression profiles.Fig. S9. ACD generates progenies with strengthened memory signatures.Table S1. Raw data files.References (47–55)

REFERENCES AND NOTES 1. S. M. Kaech, E. J. Wherry, Heterogeneity and cell-fate decisions in effector and memory

CD8+ T cell differentiation during viral infection. Immunity 27, 393–405 (2007). 2. S. M. Kaech, W. Cui, Transcriptional control of effector and memory CD8+ T cell

differentiation. Nat. Rev. Immunol. 12, 749–761 (2012). 3. E. L. Pearce, M. C. Walsh, P. J. Cejas, G. M. Harms, H. Shen, L. S. Wang, R. G. Jones, Y. Choi,

Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460, 103–107 (2009).

4. D. J. van der Windt, E. M. Dons, C. L. Montoya, M. Ezzelarab, C. Long, R. F. Wolf, J. N. M. IJzermans, F. G. Lakkis, D. K. C. Cooper, T-lymphocyte homeostasis and function in infant baboons: Implications for transplantation. Transpl. Int. 25, 218–228 (2012).

5. L. V. Sinclair, J. Rolf, E. Emslie, Y.-B. Shi, P. M. Taylor, D. A. Cantrell, Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat. Immunol. 14, 500–508 (2013).

6. M. D. Buck, D. O’Sullivan, R. I. Klein Geltink, J. D. Curtis, C.-H. Chang, D. E. Sanin, J. Qiu, O. Kretz, D. Braas, G. J. W. van der Windt, Q. Chen, S. C.-C. Huang, C. M. O’Neill, B. T. Edelson, E. J. Pearce, H. Sesaki, T. B. Huber, A. S. Rambold, E. L. Pearce, Mitochondrial dynamics controls T cell fate through metabolic programming. Cell 166, 63–76 (2016).

7. M. D. Buck, R. T. Sowell, S. M. Kaech, E. L. Pearce, Metabolic instruction of immunity. Cell 169, 570–586 (2017).

8. L. Pace, C. Goudot, E. Zueva, P. Gueguen, N. Burgdorf, J. J. Waterfall, J. P. Quivy, G. Almouzni, S. Amigorena, The epigenetic control of stemness in CD8+ T cell fate commitment. Science 359, 177–186 (2018).

9. B. Youngblood, J. S. Hale, H. T. Kissick, E. Ahn, X. Xu, A. Wieland, K. Araki, E. E. West, H. E. Ghoneim, Y. Fan, P. Dogra, C. W. Davis, B. T. Konieczny, R. Antia, X. Cheng, R. Ahmed, Effector CD8 T cells dedifferentiate into long-lived memory cells. Nature 552, 404–409 (2017).

10. J. T. Chang, M. L. Ciocca, I. Kinjyo, V. R. Palanivel, C. E. McClurkin, C. S. DeJong, E. C. Mooney, J. S. Kim, N. C. Steinel, J. Oliaro, C. C. Yin, B. I. Florea, H. S. Overkleeft, L. J. Berg, S. M. Russell, G. A. Koretzky, M. S. Jordan, S. L. Reiner, Asymmetric proteasome segregation as a mechanism for unequal partitioning of the transcription factor T-bet during T lymphocyte division. Immunity 34, 492–504 (2011).

11. J. T. Chang, V. R. Palanivel, I. Kinjyo, F. Schambach, A. M. Intlekofer, A. Banerjee, S. A. Longworth, K. E. Vinup, P. Mrass, J. Oliaro, N. Killeen, J. S. Orange, S. M. Russell, W. Weninger, S. L. Reiner, Asymmetric T lymphocyte division in the initiation of adaptive immune responses. Science 315, 1687–1691 (2007).

12. J. Oliaro, V. van Ham, F. Sacirbegovic, A. Pasam, Z. Bomzon, K. Pham, M. J. Ludford-Menting, N. J. Waterhouse, M. Bots, E. D. Hawkins, S. V. Watt, L. A. Cluse, C. J. P. Clarke, D. J. Izon, J. T. Chang, N. Thompson, M. Gu, R. W. Johnstone, M. J. Smyth, P. O. Humbert, S. L. Reiner, S. M. Russell, Asymmetric cell division of T cells upon antigen presentation uses multiple conserved mechanisms. J. Immunol. 185, 367–375 (2010).

13. K. N. Pollizzi, I.-H. Sun, C. H. Patel, Y.-C. Lo, M.-H. Oh, A. T. Waickman, A. J. Tam, R. L. Blosser, J. Wen, G. M. Delgoffe, J. D. Powell, Asymmetric inheritance of mTORC1 kinase activity during division dictates CD8+ T cell differentiation. Nat. Immunol. 17, 704–711 (2016).

14. K. C. Verbist, C. S. Guy, S. Milasta, S. Liedmann, M. M. Kamiński, R. Wang, D. R. Green, Metabolic maintenance of cell asymmetry following division in activated T lymphocytes. Nature 532, 389–393 (2016).

15. M. L. Dustin, S. K. Bromley, Z. Kan, D. A. Peterson, E. R. Unanue, Antigen receptor engagement delivers a stop signal to migrating T lymphocytes. Proc. Natl. Acad. Sci. U.S.A. 94, 3909–3913 (1997).

16. V. Das, B. Nal, A. Dujeancourt, M. I. Thoulouze, T. Galli, P. Roux, A. Dautry-Varsat, A. Alcover, Activation-induced polarized recycling targets T cell antigen receptors to the immunological synapse; involvement of SNARE complexes. Immunity 20, 577–588 (2004).

17. B. Kakaradov, J. Arsenio, C. E. Widjaja, Z. He, S. Aigner, P. J. Metz, B. Yu, E. J. Wehrens, J. Lopez, S. H. Kim, E. I. Zuniga, A. W. Goldrath, J. T. Chang, G. W. Yeo, Early transcriptional and epigenetic regulation of CD8+ T cell differentiation revealed by single-cell RNA sequencing. Nat. Immunol. 18, 422–432 (2017).

18. C. G. King, S. Koehli, B. Hausmann, M. Schmaler, D. Zehn, E. Palmer, T cell affinity regulates asymmetric division, effector cell differentiation, and tissue pathology. Immunity 37, 709–720 (2012).

19. M. L. Ciocca, B. E. Barnett, J. K. Burkhardt, J. T. Chang, S. L. Reiner, Cutting edge: Asymmetric memory T cell division in response to rechallenge. J. Immunol. 188, 4145–4148 (2012).

20. S. D. Blackburn, H. Shin, G. J. Freeman, E. J. Wherry, Selective expansion of a subset of exhausted CD8 T cells by alphaPD-L1 blockade. Proc. Natl. Acad. Sci. U.S.A. 105, 15016–15021 (2008).

21. K. Araki, A. P. Turner, V. O. Shaffer, S. Gangappa, S. A. Keller, M. F. Bachmann, C. P. Larsen, R. Ahmed, mTOR regulates memory CD8 T-cell differentiation. Nature 460, 108–112 (2009).

22. L. Zhang, B. O. Tschumi, I. C. Lopez-Mejia, S. G. Oberle, M. Meyer, G. Samson, M. A. Rüegg, M. N. Hall, L. Fajas, D. Zehn, J. P. Mach, A. Donda, P. Romero, Mammalian target of rapamycin complex 2 controls CD8 T cell memory differentiation in a Foxo1-dependent manner. Cell Rep. 14, 1206–1217 (2016).

23. S. Baldi, A. Bolognesi, A. C. Meinema, Y. Barral, Heat stress promotes longevity in budding yeast by relaxing the confinement of age-promoting factors in the mother cell. eLife 6, e28329 (2017).

24. K. N. Pollizzi, J. D. Powell, Regulation of T cells by mTOR: The known knowns and the known unknowns. Trends Immunol. 36, 13–20 (2015).

25. R. Zoncu, A. Efeyan, D. M. Sabatini, mTOR: From growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 12, 21–35 (2011).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 15: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

Borsa et al., Sci. Immunol. 4, eaav1730 (2019) 12 April 2019

S C I E N C E I M M U N O L O G Y | R E S E A R C H A R T I C L E

15 of 15

26. D. T. Utzschneider, M. Charmoy, V. Chennupati, L. Pousse, D. P. Ferreira, S. Calderon-Copete, M. Danilo, F. Alfei, M. Hofmann, D. Wieland, S. Pradervand, R. Thimme, D. Zehn, W. Held, T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

27. S. Mandala, R. Hajdu, J. Bergstrom, E. Quackenbush, J. Xie, J. Milligan, R. Thornton, G.-J. Shei, D. Card, C. Keohane, M. Rosenbach, J. Hale, C.-L. Lynch, K. Rupprecht, W. Parsons, H. Rosen, Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 296, 346–349 (2002).

28. M. Matloubian, C. G. Lo, G. Cinamon, M. J. Lesneski, Y. Xu, V. Brinkmann, M. L. Allende, R. L. Proia, J. G. Cyster, Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427, 355–360 (2004).

29. E. V. Berdyshev, I. Gorshkova, A. Skobeleva, R. Bittman, X. Lu, S. M. Dudek, T. Mirzapoiazova, J. G. N. Garcia, V. Natarajan, FTY720 inhibits ceramide synthases and up-regulates dihydrosphingosine 1-phosphate formation in human lung endothelial cells. J. Biol. Chem. 284, 5467–5477 (2009).

30. P. J. Metz, J. Arsenio, B. Kakaradov, S. H. Kim, K. A. Remedios, K. Oakley, K. Akimoto, S. Ohno, G. W. Yeo, J. T. Chang, Regulation of asymmetric division and CD8+T lymphocyte fate specification by protein kinase C and protein kinase C/. J. Immunol. 194, 2249–2259 (2015).

31. A. M. Butler, M. L. Scotti Buzhardt, E. Erdogan, S. Li, K. S. Inman, A. P. Fields, N. R. Murray, A small molecule inhibitor of atypical protein kinase C signaling inhibits pancreatic cancer cell transformed growth and invasion. Oncotarget 6, 15297–15310 (2015).

32. M. Stallings-Mann, L. Jamieson, R. P. Regala, C. Weems, N. R. Murray, A. P. Fields, A novel small-molecule inhibitor of protein kinase Ciota blocks transformed growth of non-small-cell lung cancer cells. Cancer Res. 66, 1767–1774 (2006).

33. B. He, S. Xing, C. Chen, P. Gao, L. Teng, Q. Shan, J. A. Gullicksrud, M. D. Martin, S. Yu, J. T. Harty, V. P. Badovinac, K. Tan, H.-H. Xue, CD8+ T cells utilize highly dynamic enhancer repertoires and regulatory circuitry in response to infections. Immunity 45, 1341–1354 (2016).

34. C. Gerlach, J. W. J. van Heijst, E. Swart, D. Sie, N. Armstrong, R. M. Kerkhoven, D. Zehn, M. J. Bevan, K. Schepers, T. N. M. Schumacher, One naive T cell, multiple fates in CD8+ T cell differentiation. J. Exp. Med. 207, 1235–1246 (2010).

35. C. Stemberger, K. M. Huster, M. Koffler, F. Anderl, M. Schiemann, H. Wagner, D. H. Busch, A single naive CD8+ T cell precursor can develop into diverse effector and memory subsets. Immunity 27, 985–997 (2007).

36. C. Stemberger, M. Neuenhahn, V. R. Buchholz, D. H. Busch, Origin of CD8+ effector and memory T cell subsets. Cell. Mol. Immunol. 4, 399–405 (2007).

37. J. Arsenio, B. Kakaradov, P. J. Metz, S. H. Kim, G. W. Yeo, J. T. Chang, Early specification of CD8+ T lymphocyte fates during adaptive immunity revealed by single-cell gene-expression analyses. Nat. Immunol. 15, 365–372 (2014).

38. J. Arsenio, P. J. Metz, J. T. Chang, Asymmetric cell division in T lymphocyte fate diversification. Trends Immunol. 36, 670–683 (2015).

39. M. M. Kaminski, S. Liedmann, S. Milasta, D. R. Green, Polarization and asymmetry in T cell metabolism. Semin. Immunol. 28, 525–534 (2016).

40. S. L. Reiner, W. C. Adams, Lymphocyte fate specification as a deterministic but highly plastic process. Nat. Rev. Immunol. 14, 699–704 (2014).

41. J. D. Powell, K. N. Pollizzi, E. B. Heikamp, M. R. Horton, Regulation of immune responses by mTOR. Annu. Rev. Immunol. 30, 39–68 (2012).

42. G. M. Delgoffe, J. D. Powell, Exploring functional in vivo consequences of the selective genetic ablation of mTOR signaling in T helper lymphocytes. Methods Mol. Biol. 821, 317–327 (2012).

43. D. Finlay, D. A. Cantrell, Metabolism, migration and memory in cytotoxic T cells. Nat. Rev. Immunol. 11, 109–117 (2011).

44. D. K. Finlay, E. Rosenzweig, L. V. Sinclair, C. Feijoo-Carnero, J. L. Hukelmann, J. Rolf, A. A. Panteleyev, K. Okkenhaug, D. A. Cantrell, PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 209, 2441–2453 (2012).

45. M. A. Cox, S. R. Barnum, D. C. Bullard, A. J. Zajac, ICAM-1-dependent tuning of memory CD8 T-cell responses following acute infection. Proc. Natl. Acad. Sci. U.S.A. 110, 1416–1421 (2013).

46. A. Scholer, S. Hugues, A. Boissonnas, L. Fetler, S. Amigorena, Intercellular adhesion molecule-1-dependent stable interactions between T cells and dendritic cells determine CD8+ T cell memory. Immunity 28, 258–270 (2008).

47. H. Pircher, P. Ohashi, G. Miescher, R. Lang, A. Zikopoulos, K. Bürki, T. W. Mak, H. Robson MacDonald, H. Hengartner, T cell receptor (TcR) beta chain transgenic mice: Studies on allelic exclusion and on the TcR+ gamma/delta population. Eur. J. Immunol. 20, 417–424 (1990).

48. C. F. Bentzinger, K. Romanino, D. Cloëtta, S. Lin, J. B. Mascarenhas, F. Oliveri, J. Xia, E. Casanova, C. F. Costa, M. Brink, F. Zorzato, M. N. Hall, M. A. Rüegg, Skeletal muscle-specific ablation of raptor, but not of rictor, causes metabolic changes and results in muscle dystrophy. Cell Metab. 8, 411–424 (2008).

49. S. Sawada, J. D. Scarborough, N. Killeen, D. R. Littman, A lineage-specific transcriptional silencer regulates CD4 gene expression during T lymphocyte development. Cell 77, 917–929 (1994).

50. R. Ahmed, A. Salmi, L. D. Butler, J. M. Chiller, M. B. Oldstone, Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J. Exp. Med. 160, 521–540 (1984).

51. M. Battegay, S. Cooper, A. Althage, J. Bänziger, H. Hengartner, R. M. Zinkernagel, Quantification of lymphocytic choriomeningitis virus with an immunological focus assay in 24- or 96-well plates. J. Virol. Methods 33, 191–198 (1991).

52. Y. Liao, G. K. Smyth, W. Shi, The Subread aligner: Fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013).

53. Y. Liao, G. K. Smyth, W. Shi, featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

54. M. D. Robinson, D. J. McCarthy, G. K. Smyth, edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

55. Q.-G. Steiner, L. A. Otten, M. J. Hicks, G. Kaya, F. Grosjean, E. Saeuberli, C. Lavanchy, F. Beermann, K. L. McClain, H. Acha-Orbea, In vivo transformation of mouse conventional CD8+ dendritic cells leads to progressive multisystem histiocytosis. Blood 111, 2073–2082 (2008).

Acknowledgments: We thank N. Oetiker and F. Wagen for excellent technical assistance and P. Romero for provision of the CD4-cre Raptorfl/fl and CD4-cre Rictorfl/fl mice. We thank members of the Oxenius, Joller, Barral, and Jessberger groups for helpful discussions. Funding: This work was supported by ETH Zurich, the Swiss National Science Foundation (grant no. 310030_166078 to A.O.; grant nos. 310030_53074 and 310030B_173338 to A.H.), and the Novartis Foundation for Biomedical Research. Author contributions: M.B., Y.B., and A.O. designed the experiments; M.B., I.B., N.S.B., K.P., F.G., N.B., and A.H. performed the experiments; RNA sequencing data analysis was carried out by A.Y., M.B., and I.S.; M.B. and A.O. analyzed the experiments; and M.B. and A.O. wrote the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: Raw sequencing data can be found at ArrayExpress (E-MTAB-7651).

Submitted 21 August 2018Accepted 19 February 2019Published 12 April 201910.1126/sciimmunol.aav1730

Citation: M. Borsa, I. Barnstorf, N. S. Baumann, K. Pallmer, A. Yermanos, F. Gräbnitz, N. Barandun, A. Hausmann, I. Sandu, Y. Barral, A. Oxenius, Modulation of asymmetric cell division as a mechanism to boost CD8+ T cell memory. Sci. Immunol. 4, eaav1730 (2019).

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from

Page 16: T CELLS Copyright © 2019 Modulation of asymmetric cell division … · Borsa et al., ci. mmunol. 4, eaav1730 2019 12 April 2019 SCIENCE IMMUNOLOGY| RESEARCH ARTICLE 1 of 15 T CELLS

T cell memory+Modulation of asymmetric cell division as a mechanism to boost CD8

Barandun, Annika Hausmann, Ioana Sandu, Yves Barral and Annette OxeniusMariana Borsa, Isabel Barnstorf, Nicolas S. Baumann, Katharina Pallmer, Alexander Yermanos, Fabienne Gräbnitz, Niculò

DOI: 10.1126/sciimmunol.aav1730, eaav1730.4Sci. Immunol. 

immunotherapies.centric−Developing methods to enhance T cell memory remains critical on several fronts, from vaccination to T cell

T cells and their ability to undergo ACD. +mTOR pathway using rapamycin enhances the memory potential of CD8developed a strategy to enhance the ability of memory T cells to undergo ACD. They show that transient inhibition of the

. haveet almemory cells in the immune system including memory T cells have the ability to undergo ACD. Borsa cells,of the daughter cells to retain stemness, whereas the other daughter cell becomes more differentiated. Akin to stem

One interesting property of stem cells is their ability to undergo asymmetric cell division (ACD), which allows oneRAPping their way to stemness

ARTICLE TOOLS http://immunology.sciencemag.org/content/4/34/eaav1730

MATERIALSSUPPLEMENTARY http://immunology.sciencemag.org/content/suppl/2019/04/08/4.34.eaav1730.DC1

REFERENCES

http://immunology.sciencemag.org/content/4/34/eaav1730#BIBLThis article cites 55 articles, 15 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.Science ImmunologyNew York Avenue NW, Washington, DC 20005. The title (ISSN 2470-9468) is published by the American Association for the Advancement of Science, 1200Science Immunology

Science. No claim to original U.S. Government WorksCopyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

by guest on May 23, 2021

http://imm

unology.sciencemag.org/

Dow

nloaded from