supplementary materials for - science · collected and sterile filtered through 0.22 μm pore...

20
www.sciencemag.org/cgi/content/full/science.1260668/DC1 Supplementary Materials for Functional heterogeneity of human memory CD4 + T cell clones primed by pathogens or vaccines Simone Becattini, Daniela Latorre, Federico Mele, Mathilde Foglierini, Corinne De Gregorio, Antonino Cassotta, Blanca Fernandez, Sander Kelderman, Ton N. Schumacher, Davide Corti, Antonio Lanzavecchia, Federica Sallusto* *Corresponding author. E-mail: [email protected] Published 4 December 2014 on Science Express DOI: 10.1126/science.1260668 This PDF file includes: Materials and Methods Figs. S1 to S7 Tables S1 and S2 References and Notes Other supplementary material for this manuscript includes the following: Data files (TCR sequences in zipped folder)

Upload: others

Post on 18-Oct-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

www.sciencemag.org/cgi/content/full/science.1260668/DC1

Supplementary Materials for

Functional heterogeneity of human memory CD4+ T cell clones primed

by pathogens or vaccines

Simone Becattini, Daniela Latorre,

Federico Mele, Mathilde Foglierini, Corinne De Gregorio,

Antonino Cassotta, Blanca Fernandez, Sander Kelderman, Ton N. Schumacher, Davide Corti,

Antonio Lanzavecchia, Federica Sallusto*

*Corresponding author. E-mail: [email protected]

Published 4 December 2014 on Science Express

DOI: 10.1126/science.1260668

This PDF file includes:

Materials and Methods

Figs. S1 to S7

Tables S1 and S2

References and Notes

Other supplementary material for this manuscript includes the following:

Data files (TCR sequences in zipped folder)

Page 2: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

2

Materials and Methods

Cells and cell sorting

Blood from healthy donors was obtained from the Swiss Blood Donation Center of

Basel and Lugano and used in compliance with the Federal Office of Public Health

(authorization no. A000197/2 to F.S). Peripheral blood mononuclear cells (PBMCs) were

isolated with Ficoll-Paque Plus (GE Healthcare). Monocytes and total CD4 T cells were

isolated by positive selection using CD14 and CD4 magnetic microbeads, respectively

(Miltenyi Biotech). Memory TH cell subsets were sorted to over 97% purity as follows

and after gating on CD8–CD14

–CD16

–CD19

–CD25

–CD56

–CD45RA

– cells:

CXCR3+CCR4

–CCR6

– cells (defined as TH1), CCR4

+CXCR3

–CCR6

– (defined as TH2),

CCR6+CXCR3

+CCR4

– (defined as TH1*), CCR6

+CCR4

+CXCR3

– (defined as TH17).

Naïve T cells were sorted as CD45RA+CCR7

+CD8

–CD14

–CD16

–CD19

–CD25

–CD56

CD45RO–CD95

–. The following fluorochrome-labeled mouse monoclonal antibodies

were used for staining: CD45RA-FITC (ALB11), CD45RO-PE (UCHL1), CD8-PE-Cy5

(B9.11), CD14-PE-Cy5 (RMO52), CD16-PE-Cy5 (3G8), CD19-PE-Cy5 (J3-119), CD25-

PE-Cy5 (B1.49.9), CD56-PE-Cy5 (N901) (from Beckman Coulter), CCR6-PE (11A9),

CCR4-PE-Cy7 (1G1) (from BD Biosciences), CXCR3-AlexaFluor 647 (G025H7),

CCR7-BV421 (G043H7) (from Biolegend), CD95-PerCP-eFluor 710 (DX2) (from

eBioscience). Cells were stained on ice for 15-20 minutes and sorted with FACSAria III

(BD Biosciences). Viable IL-17+ (IFN-

– IL-4

–), IFN-

+ (IL-4

– IL-17

–), and IL-4

+ (IFN-

IL-17–) cells were FACS-sorted using the cytokine secretion assay (Miltenyi Biotec).

Briefly, primed T cells were restimulated for 3 hours with PMA and Ionomycin, washed

in cold buffer and incubated with a mix of IFN-γ, IL-4 and IL-17 catch reagents for 5

minutes on ice. Warm medium was added and cells were incubated for 45 minutes at

37°C under slow continuous rotation for cytokine secretion. Cells were then washed and

stained with a cytokine-detection antibody mix containing IFN-γ-FITC, IL-4-PE, IL-17-

APC detection antibodies for 10 minutes on ice.

Ex-vivo T cell stimulation and intracellular staining

T cells were cultured in RPMI 1640 supplemented with 2 mM glutamine, 1% non-

essential amino acids, 1% sodium pyruvate, 1% penicillin/streptomycin (all from Life

Technologies), and 5% human serum (Swiss Red Cross). For some experiments, up to

500 IU/ml IL-2 was added to the medium. Sorted T cells were labeled with

carboxyfluorescein succinimidyl ester (CFSE) or Cell Trace violet (CTV) and cultured at

a ratio of 2:1 with irradiated autologous monocytes pre-pulsed for 3-5h with the antigen

of interest. In the case of C. albicans, cells were stimulated with a mixture of heat-killed

particles and lysate. Proliferating cells were sorted on day 6 (memory) or day 15 (naïve)

based on CFSE or CTV dilution and immediately stained for analysis of surface markers

or stimulated with PMA and ionomycin for 5h in the presence of brefeldin A for the last

2.5 h (all reagents from Sigma-Aldrich). Cells were fixed and permeabilized with

Cytofix/Cytoperm (BD Biosciences), according to the manufacturer’s instructions, and

then stained with the following anti-cytokine antibodies: IL-17A-eFluor660 (64DEC17),

IL-22-PerCP-eFluor710 (22URTI) (eBioscience), IFN- -APC-Cy7 (4S.B3) (Biolegend),

IFN- -FITC (B27), IL-4-PE (MP425D2) (BD Biosciences). Cytokine concentration in the

culture supernatants was assessed using the FlowCytomix assay (eBioscience), according

to manufacturer’s instruction.

Page 3: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

3

Microbes and antigens

C. albicans strains SC5314 or ATTC 14053 were used. C. albicans was cultured in

YPD medium for 16 hours at 30°C, extensively washed in PBS and heat-inactivated at

65°C for 30 minutes. Ratio used for stimulation assays was three particles per monocyte.

Lysate was prepared from mixed cultures of conidia and hyphae. Briefly, C. albicans was

cultured in YPD medium for 16 hours at 30°C or in YPD supplemented with 10% human

serum for 16 hours at 37°C. The resulting cultures yielded almost pure populations of

conidia and hyphae, respectively. Cells from the two cultures were extensively washed,

resuspended in PBS with complete protease inhibitor cocktail (Roche), mixed and

sonicated on ice for 30 consecutive cycles (45 seconds on, 90 seconds off, amplitude =

100). The suspension was then centrifuged at max speed for 15 minutes, and supernatant

collected and sterile filtered through 0.22 μm pore membranes. Quantification was

performed using Bradford reagent (Bio-Rad). C. albicans lysate was used at a

concentration of 2.5 μg/ml, M. tuberculosis lysate (strain H37Rv, from Bei Resources)

and Tetanus Toxoid (from Novartis Vaccines, Siena, Italy) were used at 5 μg/ml.

Amplification of TCR genes

Individual T cell clone total cDNA was obtained from 103 – 10

4 cells/reaction.

Reaction was carried out using oligo dT(15) primers (Promega) and Superscript III (Life

Technologies) reverse transcriptase, in a reaction mix containing DTT, NP40, dNTPs,

RNAsin (Promega). Reactions were run with the following conditions: 42°C x 10

minutes, 25°C x 10 minutes, 50°C x 1 hour, 94°C x 5 minutes. T cell receptor (TCR)

sequences from T cells were identified from cDNA (40). DNA libraries were prepared

from the cDNA using the Illumina TruSeq DNA library preparation kit. The resulting

DNA libraries were sequenced on an Illumina MiSeq sequenzer using Paired-end 150bp

chemistry. Sequencing reads in FASTQ files were mapped to the human genome, build

NCBI36/hg18, using BWA and SAMtools (41, 42). PCR duplicates in resulting BAM

files were filtered using Picard (http://picard.sourceforge.net). CDR3 TCR sequences

were identified as previously reported (43). TCRα and β sequences were inferred using

an unpublished python script developed by NKI-AVL. In addition, TCRα and β

sequences were performed at the IRB. Three μl of cDNA were added to a PCR mix (final

volume 25 μl) containing PfuUltra II Fusion HS DNA Polymerase (Agilent Genomics).

Sequences were amplified using one or both the designed TCR Vα or Vβ-specific

forward primer pools (pool fw1, pool fw2) and TAC-rev or TBC-rev reverse primers

pairing to α chain constant region or C1-C2 β chain constant region, respectively. PCR

reactions were performed with the following conditions: 95°C x 1 minute; (95°C x 20

seconds; 50°C x 20 seconds; 72°C x 30 seconds) x 45 cycles; 72°C x 3 minutes.

Sequence amplification was assessed through agarose gel electrophoresis; successfully

amplified fragments were sequenced through Sanger method using TAC-rev or TBC-rev

primer.

RNA extraction and qRT-PCR

Total RNA was extracted using TRIzol reagent (Life Technologies) or E.Z.N.A.

DNA/RNA Isolation Kit (Omega Bio-tek) according to manufacturer’s instructions.

QScript cDNA SuperMix (Quanta Biosciences) was used for cDNA synthesis.

Transcripts were quantified by qRT–PCR on ABI PRISM 7900HT, with predesigned

Page 4: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

4

TaqMan Gene Expression Assays: RORC (Hs01076122_m1), TBX21 (Hs00203436_m1),

GATA3 (Hs00231122_m1, all from Life Technologies Applied Biosystems). Reactions

were run on a 7900 Fast Real Time PCR System (Life Technologies Applied

Biosystems). Expression of target genes was normalized to 18S ribosomal RNA or TBP

RNA and expressed as arbitrary units (A.U.).

TCRβ deep sequencing

Antigen-specific T cells were obtained as described above. A minimum number of

106 cells were obtained for all experiments, and each sample was split in two, half of

which was frozen as a backup. In case cells sorted upon stimulation did not reach the

required number, they were expanded for 1 to 5 days in the presence of 50 IU/ml IL-2.

Cells to be analyzed by deep sequencing were centrifuged and washed in PBS, and

genomic DNA was extracted from the pellet using QUIAamp Micro Kit (Qiagen),

according to manufacturer’s instruction. Genomic DNA quantity and purity were

assessed through spectrophotometric analysis. Deep sequencing of TCRβ was performed

by Adaptive Biotechnologies Corp. (Seattle, WA) using the ImmunoSEQ assay

(http://www.immunoseq.com). Briefly, following multiplex PCR reaction designed to

target any TCRβ CDR3 fragments, amplicons were sequenced using the Illumina HiSeq

platform. Raw data consisting of all retrieved sequences of 87 nucleotides or

corresponding amino acidic sequences and containing CDR3 region were exported and

further processed. If not differently stated in the text, the assay was performed at survey

level (detection sensitivity, 1 cell in 40,000); for some samples, a deep level analysis was

used (detection sensitivity, 1 cell in 200,000).

TCRβ sequence analysis

Data sets of TCRβ sequences were analyzed using algorithms written in Java. DNA

sequences containing frame-shift or stop codons were removed prior to analysis. The

experimental noise was determined for each batch of experiments with parallel analysis

of antigen-specific memory T cells (obtained as described above) that were split in two

before DNA extraction and sequencing. For each control, all the sequences (shared and

not shared) were plotted together. The value of the non-shared sequence with the highest

number of reads was set as threshold. Sequences with a value of reads below threshold

level were deleted from the experiment. The percentage of shared clonotypes was

calculated using the Jaccard index [J=(A∩B)/(A∪B)] as number of shared clonotypes

between two subsets divided by the total number of clonotypes present in the same

subsets, and normalized by 100. The percentage of shared reads was calculated as

average of the cumulative frequencies of the shared clonotypes in each of the two

subsets. For instance, if shared clonotypes within the A and B subsets accounted for 30%

and 40% of the total populations, respectively, based on their read count, the size of the

shared population would be calculated as 35% of the A-B compartment. A disparity

index was calculated as previously described (44) as:

Where fi represents the number of reads of the clonotype i divided by the total number of

reads in the sample, and N is the total number of clonotypes present in the same sample.

Page 5: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

5

The index ranges from D=0 and D=1, D=0 representing a population composed by

equally expanded clonotypes and D=1 a population where only one clonotype dominates.

Statistical analysis

Statistical analysis was performed with the Prism software (GraphPad). Data in

columns represent means ± SEM values, and significance was assessed by non-

parametric paired Friedman test. Non-parametric Spearman correlation coefficient was

determined for shared sequences.

Page 6: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

6

Fig. S1.

Sorting strategy to identify functional memory CD4 T cell subsets. (A and B)

Expression of chemokine receptors identifies four subsets of human memory CD4 T

cells. Cells expressing CD45RA, CD25, CD14, CD19 were excluded from the gate.

Representative dot plot in one donor (A) and percentage of each subset relative to total

memory CD4 T cells in thirteen donors (B). Data are means ± SEM. (C and D) Cytokine

production by the four memory subsets following stimulation with PMA and Ionomycin.

Dot plots are from a representative donor (C). Means ± SEM of four donors (D). (E)

TBX21, GATA3, and RORC mRNA levels in the four memory subsets analyzed

immediately after sorting. Data are the means ± SEM of four donors.

Page 7: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

7

Fig. S2

C. albicans–specific clonotype sharing between samples that were split before or

after antigenic stimulation. (A) 1.5x106 TH17 cells were stimulated with C. albicans.

After 6 days, CFSElo

cells were sorted and the sample was divided in two halves that

were separately subjected to genomic DNA extraction and sequencing. Shown are

absolute numbers of reads. The total number of clonotypes in the two samples is

indicated on x and y axis. Values in the lower right corner represent the number of

clonotypes and percentage of reads shared between the two samples. The Spearman

correlation and paired t test value are shown in the upper part. (B and C) Sample split

control experiments were performed for the different donors analyzed and the highest

read value at which a sequence was retrieved in only one of the two split samples was set

as a cutoff. Sequences with read values below the cutoff were removed. Shown in (B) are

the removed sequences (expressed as percentage of total reads) in all analyzed donors (C.

albicans, n = 5; TT n = 4; every dot represents a donor). Shown in (C) is the comparison

between sizes of validated versus removed (below-threshold) sequences, expressed as

average number of reads; every dot represents averaging from one donor. (D) 1.5x106

TH17 cells were sorted and divided in two wells that were stimulated with C. albicans in

the presence of autologous monocytes. TCRβ analysis was performed on day 6 on

genomic DNA from CFSElo

cells that had proliferated independently in the two cultures.

Shown is the number of clonotypes detected in each well, the number of clonotypes and

percent of reads shared between the two wells. Shown is also the Spearman correlation

and paired t test value. Data are representative of three separate experiments performed

with C. albicans and one experiment performed with the four memory subsets (TH1, TH2,

TH1*, and TH17) and TT as antigen.

Page 8: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

8

Fig. S3

Clonotypes shared among subsets of C. albicans–specific T cells. Venn diagrams show

for four donors the number of unique and shared clonotypes among C. albicans–specific

T cells isolated from the four memory subsets.

Donor CA-01

5

31 782

24

124

110 171

462

8

17

26

724

6

Th1

Th2 Th1*

Th17

Donor CA-03

4

17 2559

95

433

320 842

1072

2

3

57

1938

10

Th1

Th2 Th1*

Th17

Donor CA-04

11

57 10529

46

160

623 142

702

15

23

43

4050

9

Th1

Th2 Th1*

Th17

Donor CA-05

28

62 22520

224

949

737 1000

1598

11

41

105

18146

23

Th1

Th2 Th1*

Th17

Page 9: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

9

Fig. S4

Sorting strategy of CCR6+ memory CD4 T cell subsets. Three subsets of CCR6

+

memory CD4 T cells were sorted according to the expression of CXCR3 and CCR4. (A)

Cytokine production by the indicated memory subsets after sorting from PBMCs and in

vitro stimulation with PMA and Ionomycin for 5 hours. (B) TBX21, GATA3, and RORC

mRNA levels in the indicated memory subsets analyzed immediately after sorting. Data

are the means ± SEM of three donors.

Page 10: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

10

Fig. S5

Clonotypic analysis of M. tuberculosis–specific CD4 T cell subsets. (A) CFSE profiles

and percentage of CFSElo

proliferating cells in memory T cell subsets stimulated with M.

tuberculosis in donor MT-01. (B) Cytokine production by CFSElo

cells measured by

intracellular cytokine staining after stimulation for 5 hours with PMA and Ionomycin.

(C) Comparison of clonotype frequency distribution in samples of T cells isolated from

M. tuberculosis–stimulated T cell subsets from donors MT-01 and MT-02. Frequencies

are shown as percentage of total reads. The total number of clonotypes in each sample is

indicated in parenthesis on x axis and y axis. Shown is the number of clonotypes shared

between the two samples; Spearman correlation and paired t test values are shown when

significant. (D) Bar histograms showing for the two donors percentage of clonotypes

(upper panel) and percentage of reads (lower panel) that are shared by the indicated

subsets.

Page 11: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

11

Fig. S6

Clonotypes shared among subsets of TT–specific T cells. Venn diagrams show for four

donors the number of unique and shared clonotypes among TT–specific T cells isolated

from the four memory subsets.

5315 5

Donor TT-01

28

106 639

32

293

262 169

161

83

30

77

Th1

Th2 Th1*

Th17

Donor TT-02

24

75 17212

152

688

818 501

536

37

65

28

4587

18

Th1

Th2 Th1*

Th17

5312 4

Donor TT-03

7

26 704

46

166

360 233

280

12

8

48

Th1

Th2 Th1*

Th17

Donor TT-04

11

16 331

77

232

101 350

214

8

11

4

2310

16

Th1

Th2 Th1*

Th17

Page 12: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

12

Fig. S7

Sorting of cytokine-secreting cells from cultures of naïve CD4 T cells primed in vitro

by C. albicans. Naïve CD4 T cells were primed in vitro by C. albicans in the presence of

autologous monocytes. On day 15, IL-17+ (IFN-

– IL-4

–), IFN-

+ (IL-17

– IL-4

–), or IL-4

+

(IL-17– IFN-

–) T cells were sorted using the cytokine secretion assay and maintained in

culture with addition of IL-2. Shown is the cytokine profile of cultured cells after 7 days.

IL-17+ (IFN-g–IL-4–) sorted T cells

IL-4+ (IFN-g–IL-17–) sorted T cells

IFN-g+ (IL-17–IL-4–) sorted T cells

15.7 22.3

24.9

36.8 0.8

3.12.2

1.345.9

3.6

0.26.9 55.3 0.7

3.2

51.2 5.3

1.8

13.9 1.6

2.9

11.9 3.2

5454.7

1.72.8

103102 104 1050

103

104

105

0

102

103

104

105

0

103

104

105

0

103

104

105

0

103

104

105

0

103

104

105

0

102

103

104

105

0

102

103

104

105

0

103

104

105

0

103102 104 1050

103102 104 1050

103 104 1050103 104 1050

103 104 1050103 104 1050

103 104 1050103 104 1050

IFN

-g

IL-4

IL-17

IL-4

IFN-gIL-17

IFN

-g

IL-4

IL-17

IL-4

IFN-gIL-17

IFN

-g

IL-4

IL-17

IL-4

IFN-gIL-17

Page 13: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

13

Table S1.

T cell counts and number of TCR clonotypes of C. albicans–specific TH cells from five

donors.

Page 14: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

14

Table S2.

TCRβ CDR3 sequences of T cell clones primed by C. albicans in vitro and sorted based

on the ability to selectively secrete the indicated cytokine. Data are from one experiment,

representative of three independent experiments performed with different donors.

Page 15: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

References and Notes

1. R. Medzhitov, Recognition of microorganisms and activation of the immune response. Nature

449, 819–826 (2007). Medline doi:10.1038/nature06246

2. F. Sallusto, A. Lanzavecchia, Heterogeneity of CD4+ memory T cells: Functional modules for

tailored immunity. Eur. J. Immunol. 39, 2076–2082 (2009). Medline

doi:10.1002/eji.200939722

3. F. P. Heinzel, M. D. Sadick, B. J. Holaday, R. L. Coffman, R. M. Locksley, Reciprocal

expression of interferon γ or interleukin 4 during the resolution or progression of murine

leishmaniasis. Evidence for expansion of distinct helper T cell subsets. J. Exp. Med. 169,

59–72 (1989). Medline doi:10.1084/jem.169.1.59

4. M. Yamamura, K. Uyemura, R. J. Deans, K. Weinberg, T. H. Rea, B. R. Bloom, R. L. Modlin,

Defining protective responses to pathogens: Cytokine profiles in leprosy lesions. Science

254, 277–279 (1991). Medline doi:10.1126/science.1925582

5. C. S. Ma, G. Y. Chew, N. Simpson, A. Priyadarshi, M. Wong, B. Grimbacher, D. A. Fulcher,

S. G. Tangye, M. C. Cook, Deficiency of Th17 cells in hyper IgE syndrome due to

mutations in STAT3. J. Exp. Med. 205, 1551–1557 (2008). Medline

doi:10.1084/jem.20080218

6. J. D. Milner, J. M. Brenchley, A. Laurence, A. F. Freeman, B. J. Hill, K. M. Elias, Y. Kanno,

C. Spalding, H. Z. Elloumi, M. L. Paulson, J. Davis, A. Hsu, A. I. Asher, J. O’Shea, S.

M. Holland, W. E. Paul, D. C. Douek, Impaired TH17 cell differentiation in subjects with

autosomal dominant hyper-IgE syndrome. Nature 452, 773–776 (2008). Medline

doi:10.1038/nature06764

7. A. Puel, S. Cypowyj, J. Bustamante, J. F. Wright, L. Liu, H. K. Lim, M. Migaud, L. Israel, M.

Chrabieh, M. Audry, M. Gumbleton, A. Toulon, C. Bodemer, J. El-Baghdadi, M.

Whitters, T. Paradis, J. Brooks, M. Collins, N. M. Wolfman, S. Al-Muhsen, M.

Galicchio, L. Abel, C. Picard, J. L. Casanova, Chronic mucocutaneous candidiasis in

humans with inborn errors of interleukin-17 immunity. Science 332, 65–68 (2011).

Medline doi:10.1126/science.1200439

Page 16: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

8. A. Iwasaki, R. Medzhitov, Regulation of adaptive immunity by the innate immune system.

Science 327, 291–295 (2010). Medline doi:10.1126/science.1183021

9. J. Zhu, H. Yamane, W. E. Paul, Differentiation of effector CD4 T cell populations. Annu. Rev.

Immunol. 28, 445–489 (2010). Medline doi:10.1146/annurev-immunol-030409-101212

10. A. O’Garra, L. Gabryšová, H. Spits, Quantitative events determine the differentiation and

function of helper T cells. Nat. Immunol. 12, 288–294 (2011). Medline

doi:10.1038/ni.2003

11. J. J. O’Shea, W. E. Paul, Mechanisms underlying lineage commitment and plasticity of

helper CD4+ T cells. Science 327, 1098–1102 (2010). Medline

doi:10.1126/science.1178334

12. L. Cosmi, R. De Palma, V. Santarlasci, L. Maggi, M. Capone, F. Frosali, G. Rodolico, V.

Querci, G. Abbate, R. Angeli, L. Berrino, M. Fambrini, M. Caproni, F. Tonelli, E.

Lazzeri, P. Parronchi, F. Liotta, E. Maggi, S. Romagnani, F. Annunziato, Human

interleukin 17-producing cells originate from a CD161+CD4+ T cell precursor. J. Exp.

Med. 205, 1903–1916 (2008). Medline doi:10.1084/jem.20080397

13. D. Bending, H. De la Peña, M. Veldhoen, J. M. Phillips, C. Uyttenhove, B. Stockinger, A.

Cooke, Highly purified Th17 cells from BDC2.5NOD mice convert into Th1-like cells in

NOD/SCID recipient mice. J. Clin. Invest. 119, 565–572 (2009). Medline

doi:10.1172/JCI37865

14. K. Hirota, J. H. Duarte, M. Veldhoen, E. Hornsby, Y. Li, D. J. Cua, H. Ahlfors, C. Wilhelm,

M. Tolaini, U. Menzel, A. Garefalaki, A. J. Potocnik, B. Stockinger, Fate mapping of IL-

17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263 (2011).

Medline doi:10.1038/ni.1993

15. S. L. Reiner, F. Sallusto, A. Lanzavecchia, Division of labor with a workforce of one:

Challenges in specifying effector and memory T cell fate. Science 317, 622–625 (2007).

Medline doi:10.1126/science.1143775

16. 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

Page 17: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

immune responses. Science 315, 1687–1691 (2007). Medline

doi:10.1126/science.1139393

17. C. Gerlach, J. W. van Heijst, E. Swart, D. Sie, N. Armstrong, R. M. Kerkhoven, D. Zehn, M.

J. Bevan, K. Schepers, T. N. Schumacher, One naive T cell, multiple fates in CD8+ T cell

differentiation. J. Exp. Med. 207, 1235–1246 (2010). Medline doi:10.1084/jem.20091175

18. 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). Medline

doi:10.1016/j.immuni.2007.10.012

19. N. J. Tubo, A. J. Pagán, J. J. Taylor, R. W. Nelson, J. L. Linehan, J. M. Ertelt, E. S. Huseby,

S. S. Way, M. K. Jenkins, Single naive CD4+ T cells from a diverse repertoire produce

different effector cell types during infection. Cell 153, 785–796 (2013). Medline

doi:10.1016/j.cell.2013.04.007

20. See supplementary materials on Science Online.

21. T cell subsets were isolated according to the differential expression of chemokine receptors,

as previously described (22). Cells in the CXCR3+CCR4–CCR6– subset (defined as TH1)

produced IFN-γ and expressed T-bet mRNA; cells in the CCR4+CXCR3–CCR6– subset

(TH2) produced IL-4 and expressed high levels of GATA3 mRNA; cells in the

CCR4+CCR6+CXCR3– subset (TH17) produced IL-17A and expressed RORγt; cells in

the CXCR3+CCR6+CCR4– (nonconventional TH1, defined as TH1*) produced IFN-γ and

low levels of IL-17A and expressed both T-bet and RORγt mRNA.

22. E. V. Acosta-Rodriguez, L. Rivino, J. Geginat, D. Jarrossay, M. Gattorno, A. Lanzavecchia,

F. Sallusto, G. Napolitani, Surface phenotype and antigenic specificity of human

interleukin 17-producing T helper memory cells. Nat. Immunol. 8, 639–646 (2007).

Medline doi:10.1038/ni1467

23. In all experiments, the specificity of proliferating memory T cells was assessed by sorting

CFSElo cells and isolating T cell clones by limiting dilution. On average, more than 95%

of the T cell clones responded to the antigen used in the initial stimulation.

Page 18: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

24. C. E. Zielinski, F. Mele, D. Aschenbrenner, D. Jarrossay, F. Ronchi, M. Gattorno, S.

Monticelli, A. Lanzavecchia, F. Sallusto, Pathogen-induced human TH17 cells produce

IFN-γ or IL-10 and are regulated by IL-1β. Nature 484, 514–518 (2012). Medline

doi:10.1038/nature10957

25. P. Bacher, C. Schink, J. Teutschbein, O. Kniemeyer, M. Assenmacher, A. A. Brakhage, A.

Scheffold, Antigen-reactive T cell enrichment for direct, high-resolution analysis of the

human naive and memory Th cell repertoire. J. Immunol. 190, 3967–3976 (2013).

Medline doi:10.4049/jimmunol.1202221

26. H. S. Robins, P. V. Campregher, S. K. Srivastava, A. Wacher, C. J. Turtle, O. Kahsai, S. R.

Riddell, E. H. Warren, C. S. Carlson, Comprehensive assessment of T-cell receptor β-

chain diversity in αβ T cells. Blood 114, 4099–4107 (2009). Medline doi:10.1182/blood-

2009-04-217604

27. T cell subsets from two healthy donors were isolated from PBMCs, labeled with CFSE, and

stimulated with M. tuberculosis. For donor MT-01, input cell numbers were TH1*, 1.5 ×

106; TH17, 1.5 × 106; CXCR3+CCR4+ TH, 1.2 × 106. For donor MT-02, input cell

numbers were TH1*, 1.5 × 106; TH17, 1.4 × 106; CXCR3+CCR4+ TH, 1.1 × 106.

Recovered CFSElo cells on day 6 for donor MT-01were TH1*, 2.2 × 106; TH17, 0.95 ×

106; CXCR3+CCR4+ TH, 2.0 × 106. For donor MT-02, these numbers were TH1*, 2.4 ×

106; TH17, 0.7 × 106; CXCR3+CCR4+ TH, 1.7 × 106.

28. C. S. Lindestam Arlehamn, A. Gerasimova, F. Mele, R. Henderson, J. Swann, J. A.

Greenbaum, Y. Kim, J. Sidney, E. A. James, R. Taplitz, D. M. McKinney, W. W. Kwok,

H. Grey, F. Sallusto, B. Peters, A. Sette, Memory T cells in latent Mycobacterium

tuberculosis infection are directed against three antigenic islands and largely contained in

a CXCR3+CCR6+ Th1 subset. PLOS Pathog. 9, e1003130 (2013). Medline

doi:10.1371/journal.ppat.1003130

29. T cell subsets from four donors were isolated and stimulated with TT. Input cell number for

each subset was 2.5 × 106 (donor TT-01), 1.5 × 106 (donors TT-02 and TT-03), and 1.1 ×

106 (donor TT-04).

Page 19: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

30. Naïve T cells were isolated from three donors. Input cell numbers were 3.2 × 106 (donor N-

01), 6.6 × 106 (donor N-02), and 8.8 × 106 (donor N-03).

31. In control experiments, the purity of the sorted populations was confirmed by measuring

IFNG, IL4, and IL17 mRNAs by qPCR immediately after sorting.

32. B. Pulendran, Variegation of the immune response with dendritic cells and pathogen

recognition receptors. J. Immunol. 174, 2457–2465 (2005). Medline

doi:10.4049/jimmunol.174.5.2457

33. H. Sigmundsdottir, E. C. Butcher, Environmental cues, dendritic cells and the programming

of tissue-selective lymphocyte trafficking. Nat. Immunol. 9, 981–987 (2008). Medline

doi:10.1038/ni.f.208

34. C. R. Plumlee, B. S. Sheridan, B. B. Cicek, L. Lefrançois, Environmental cues dictate the

fate of individual CD8+ T cells responding to infection. Immunity 39, 347–356 (2013).

Medline doi:10.1016/j.immuni.2013.07.014

35. M. E. Birnbaum, J. L. Mendoza, D. K. Sethi, S. Dong, J. Glanville, J. Dobbins, E. Ozkan, M.

M. Davis, K. W. Wucherpfennig, K. C. Garcia, Deconstructing the peptide-MHC

specificity of T cell recognition. Cell 157, 1073–1087 (2014). Medline

36. R. M. Welsh, L. K. Selin, No one is naive: The significance of heterologous T-cell immunity.

Nat. Rev. Immunol. 2, 417–426 (2002). Medline

37. D. Mason, A very high level of crossreactivity is an essential feature of the T-cell receptor.

Immunol. Today 19, 395–404 (1998). Medline doi:10.1016/S0167-5699(98)01299-7

38. L. Rivino, M. Messi, D. Jarrossay, A. Lanzavecchia, F. Sallusto, J. Geginat, Chemokine

receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among

human CD4+ central memory T cells. J. Exp. Med. 200, 725–735 (2004). Medline

doi:10.1084/jem.20040774

39. Production of cytokines was measured on day 6 in the supernatants of 5 × 104 naïve CD4 T

cells stimulated with plate-bound CD3 and CD28 antibodies. Mean values and SEM (n =

3) were as follows: IFN-γ, 414 ± 270.2; IL-17, 30.7 ± 3.3; IL-22, 391.6 ± 129.2; IL-4, 3.6

± 3.6; IL-5, 9.6 ± 5.4; IL-13, 91 ± 20.5.

Page 20: Supplementary Materials for - Science · collected and sterile filtered through 0.22 μm pore membranes. Quantification was performed using Bradford reagent (Bio-Rad). C. albicans

40. F. Tang, C. Barbacioru, E. Nordman, B. Li, N. Xu, V. I. Bashkirov, K. Lao, M. A. Surani,

RNA-Seq analysis to capture the transcriptome landscape of a single cell. Nat. Protoc. 5,

516–535 (2010). Medline doi:10.1038/nprot.2009.236

41. H. Li, R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform.

Bioinformatics 25, 1754–1760 (2009). Medline doi:10.1093/bioinformatics/btp324

42. H. Li, B. Handsaker, A. Wysoker, T. Fennell, J. Ruan, N. Homer, G. Marth, G. Abecasis, R.

Durbin, The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–

2079 (2009). Medline doi:10.1093/bioinformatics/btp352

43. D. A. Bolotin, M. Shugay, I. Z. Mamedov, E. V. Putintseva, M. A. Turchaninova, I. V.

Zvyagin, O. V. Britanova, D. M. Chudakov, MiTCR: Software for T-cell receptor

sequencing data analysis. Nat. Methods 10, 813–814 (2013). Medline

doi:10.1038/nmeth.2555

44. C. Gerlach, J. C. Rohr, L. Perié, N. van Rooij, J. W. van Heijst, A. Velds, J. Urbanus, S. H.

Naik, H. Jacobs, J. B. Beltman, R. J. de Boer, T. N. Schumacher, Heterogeneous

differentiation patterns of individual CD8+ T cells. Science 340, 635–639 (2013).

Medline doi:10.1126/science.1235487