Volume 38, Issue 4, Pages (April 2013)

Slides:



Advertisements
Similar presentations
Volume 45, Issue 1, Pages (July 2016)
Advertisements

Volume 42, Issue 2, Pages (February 2015)
Volume 45, Issue 2, Pages (August 2016)
Volume 45, Issue 5, Pages (November 2016)
Sustained Interactions between T Cell Receptors and Antigens Promote the Differentiation of CD4+ Memory T Cells  Chulwoo Kim, Theodore Wilson, Kael F.
Local Inflammatory Cues Regulate Differentiation and Persistence of CD8+ Tissue- Resident Memory T Cells  Tessa Bergsbaken, Michael J. Bevan, Pamela J.
Volume 36, Issue 6, Pages (June 2012)
Volume 27, Issue 4, Pages (October 2007)
Volume 40, Issue 6, Pages (June 2014)
Volume 13, Issue 6, Pages (November 2015)
Volume 35, Issue 3, Pages (September 2011)
Volume 31, Issue 5, Pages (November 2009)
Volume 12, Issue 12, Pages (September 2015)
Volume 23, Issue 7, Pages (May 2018)
Volume 21, Issue 7, Pages (November 2017)
Volume 11, Issue 6, Pages (June 2012)
Volume 25, Issue 3, Pages (September 2006)
Protective Capacity of Memory CD8+ T Cells Is Dictated by Antigen Exposure History and Nature of the Infection  Jeffrey C. Nolz, John T. Harty  Immunity 
Volume 33, Issue 1, Pages (July 2010)
Volume 21, Issue 1, Pages (October 2017)
Volume 45, Issue 1, Pages (July 2016)
Volume 31, Issue 5, Pages (November 2009)
Volume 29, Issue 1, Pages (July 2008)
Transforming Growth Factor-β Signaling Controls the Formation and Maintenance of Gut-Resident Memory T Cells by Regulating Migration and Retention  Nu.
Volume 35, Issue 4, Pages (October 2011)
Volume 29, Issue 6, Pages (December 2008)
Volume 28, Issue 5, Pages (May 2008)
Volume 43, Issue 4, Pages (October 2015)
Volume 38, Issue 1, Pages (January 2013)
Volume 29, Issue 1, Pages (July 2008)
Volume 35, Issue 2, Pages (August 2011)
Blimp-1 Transcription Factor Is Required for the Differentiation of Effector CD8+ T Cells and Memory Responses  Axel Kallies, Annie Xin, Gabrielle T.
Volume 13, Issue 6, Pages (November 2015)
Volume 40, Issue 5, Pages (May 2014)
An Interleukin-21- Interleukin-10-STAT3 Pathway Is Critical for Functional Maturation of Memory CD8+ T Cells  Weiguo Cui, Ying Liu, Jason S. Weinstein,
Volume 35, Issue 4, Pages (October 2011)
Volume 40, Issue 3, Pages (March 2014)
Volume 29, Issue 1, Pages (July 2008)
Opposing Effects of TGF-β and IL-15 Cytokines Control the Number of Short-Lived Effector CD8+ T Cells  Shomyseh Sanjabi, Munir M. Mosaheb, Richard A.
Volume 43, Issue 5, Pages (November 2015)
Volume 39, Issue 1, Pages (July 2013)
Volume 34, Issue 6, Pages (June 2011)
Volume 29, Issue 4, Pages (October 2008)
Volume 44, Issue 5, Pages (May 2016)
Volume 38, Issue 3, Pages (March 2013)
E3 Ubiquitin Ligase VHL Regulates Hypoxia-Inducible Factor-1α to Maintain Regulatory T Cell Stability and Suppressive Capacity  Jee H. Lee, Chris Elly,
Cell-Intrinsic IL-27 and gp130 Cytokine Receptor Signaling Regulates Virus-Specific CD4+ T Cell Responses and Viral Control during Chronic Infection 
Volume 48, Issue 4, Pages e8 (April 2018)
Matthew A. Williams, Eugene V. Ravkov, Michael J. Bevan  Immunity 
Volume 27, Issue 2, Pages (August 2007)
Susan M. Kaech, Scott Hemby, Ellen Kersh, Rafi Ahmed  Cell 
Volume 32, Issue 1, Pages (January 2010)
Courtney R. Plumlee, Brian S. Sheridan, Basak B. Cicek, Leo Lefrançois 
Volume 28, Issue 5, Pages (May 2008)
Volume 40, Issue 2, Pages (February 2014)
Volume 38, Issue 6, Pages (June 2013)
Volume 38, Issue 2, Pages (February 2013)
Volume 22, Issue 8, Pages (February 2018)
Volume 40, Issue 3, Pages (March 2014)
Volume 35, Issue 4, Pages (October 2011)
Volume 30, Issue 5, Pages (May 2009)
Volume 31, Issue 2, Pages (August 2009)
CD25 surface expression and TCR signal strength predict T helper differentiation and memory potential of early effector T cells in vivo. CD25 surface expression.
Volume 30, Issue 2, Pages (February 2009)
CD4+ memory T cells derived from either CD25hi or CD25lo effector cells respond robustly to secondary challenge. CD4+ memory T cells derived from either.
CD25 expression predicts effector and memory differentiation.
Volume 35, Issue 3, Pages (September 2011)
Volume 33, Issue 2, Pages (August 2010)
Volume 8, Issue 2, Pages (August 2010)
Volume 39, Issue 2, Pages (August 2013)
Presentation transcript:

Volume 38, Issue 4, Pages 805-817 (April 2013) Distinct Memory CD4+ T Cells with Commitment to T Follicular Helper- and T Helper 1- Cell Lineages Are Generated after Acute Viral Infection  J. Scott Hale, Ben Youngblood, Donald R. Latner, Ata Ur Rasheed Mohammed, Lilin Ye, Rama S. Akondy, Tuoqi Wu, Smita S. Iyer, Rafi Ahmed  Immunity  Volume 38, Issue 4, Pages 805-817 (April 2013) DOI: 10.1016/j.immuni.2013.02.020 Copyright © 2013 Elsevier Inc. Terms and Conditions

Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions

Figure 1 Phenotypic Heterogeneity of Virus-Specific CD4+ T Cells Is Maintained during Effector and Memory Differentiation We adoptively transferred 2 × 105 CD45.1+ LCMV-specific naive SMARTA transgenic CD4+ T cells into CD45.2+ naive recipients that were then infected with 2 × 105 PFU of LCMV Armstrong. FACS plots are gated on CD4+CD45.1+ SMARTA cells at the indicated time points relative to infection. (A) Kinetics of splenic SMARTA CD4+ T cells. (B) CXCR5, PD-1, ICOS, GL-7, Ly6c, and granzyme B analysis of naive, effector, and memory SMARTA CD4+ T cells. (C) Analysis of T-bet and Bcl6 expression. (D) Analysis of Bcl6 and CXCR5 on effector (day 8) and memory (day 100–133) SMARTA cells in blood and spleen. (E) Bcl6 MFI of CXCR5+ gated effector and memory SMARTA cells. Statistically significant p values are shown and were determined by using a two-tailed unpaired Students t test. (F) The frequency of effector and memory SMARTA cells that are CXCR5+ and CXCR5− in the following tissues are as follows: Spl, spleen; Bl, blood; iLN, inguinal lymph node; mLN, mesenteric lymph node; BM, bone marrow; Liv, liver; Lung, and IEL, intestinal intraepithelial lymphocytes. Graphs show the mean and SEM. n = 3 mice at each time point. (G) Plots of CD4+CD45.1+ gated SMARTA cells with gates indicating the Ly6c+CXCR5− (blue), CXCR5+Ly6clo (red), and CXCR5+Ly6cint (green) subsets. Chart indicates the number of each subset in spleen following infection (n ≥ 3 at each time point). Error bars represent the SEM. See also Figure S1 and Table S1. Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions

Figure 2 Transcriptional Profiling Suggests a Lineage Relationship between Tfh Effector and Memory Cells and between Th1 Effector and Memory Cells Microarray analysis of CD4+ SMARTA CXCR5−Ly6chi (blue), CXCR5+Ly6clo (red), and CXCR5+Ly6cint (green) subsets sorted from chimeric mice at effector (day 6 postinfection) and memory time points (day 68–147 p.i.). (A) The 30 most upregulated and 30 most downregulated genes in the day 6 Ly6hiCXCR5− (Th1) relative to the day 6 CXCR5+Ly6clo (Tfh) SMARTA CD4+ T cells were identified and their expression in all subsets at effector and memory stages is shown as a heatmap. (B–F) Expression of select genes as determined by microarray analysis, including (B) Cxcr5 and Ly6c, (C) transcription factors, (D) cytokine and chemokine receptors, (E) cytokines and cytotoxic effector molecules, and (F) costimulatory and inhibitory receptors. Data are shown as fold change relative to naive SMARTA CD4+ T cells. Bars show the average value for each indicated population; effector CXCR5−Ly6chi (n = 2), effector CXCR5+Ly6clo (n = 2), memory CXCR5−Ly6chi (n = 4), memory CXCR5+Ly6clo (n = 4), and memory CXCR5+Ly6cint (n = 3). See also Figure S2. Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions

Figure 3 Th1 and Tfh Memory CD4+ T Cells Are Committed for Recall of Lineage-Specific Functions CD45.1 congenically marked CXCR5−Ly6chi (blue), CXCR5+Ly6clo (red), and CXCR5+Ly6cint (green) memory CD4+ subsets (between days 56 and 101 post-infection) were FACS purified to >97%. We adoptively transferred 8 × 103 sorted cells into naive CD45.2 recipients, and recipient mice were then infected with LCMV Armstrong 16–20 hr later. In (C)–(I), the phenotype and function of transferred SMARTA cells were analyzed 7 days postinfection. (A) Representative postsort analysis of memory SMARTA subsets and cartoon of experimental setup. (B) Absolute number of transferred CD4+ CD45.1+ SMARTA splenocytes 7 days following rechallenge with LCMV. The relative fold increase of each population, assuming a 10% take of transferred SMARTA cells, is shown above each bar. (C) Representative CXCR5 and Ly6c analysis. (D) Chart shows the percent of transferred SMARTA cells that are CXCR5+ICOS+ Tfh cells. (E) The percent of SMARTA cells that are CXCR5+GL-7+ germinal center Tfh cells. (F) Bcl6 MFI of SMARTA cells. (G) T-bet MFI of SMARTA cells. (H) Chart shows the percent of B220+ gated splenic B cells that are Fas+PNA+ germinal center B cells. Data were combined from two experiments. (I) Chart shows the percent of transferred SMARTA cells that are CXCR5−granzymeBhi cells. Data in (B), (D), (E), and (I) are combined from four independent experiments for a total of n = 11–14 mice per group. Data in (F) and (G) were from a single experiment (n = 3 per group) and were representative of two to four independent experiments. Statistically significant p values are shown and were determined using a two-tailed unpaired Student’s t test. Error bars represent the SEM. See also Figure S3. Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions

Figure 4 Lineage Commitment of Th1 and Tfh Memory Subsets Is Maintained in the Absence of Antigen CD45.1 congenically marked CXCR5−Ly6chi (blue) and CXCR5+ (red) memory SMARTA CD4+ T cell subsets 34 days postinfection were FACS purified to >97%. We adoptively transferred 8 × 104 purified cells into naive CD45.2 recipients. Twenty-eight days following cell transfer, naive recipients were infected with 2 × 105 p.f.u. LCMV Armstrong 16–20 hr later. The phenotype (B–H) of transferred CD4+CD45.1+ gated SMARTA cells was analyzed 7 days postinfection. (A) Cartoon of experimental setup. (B) CXCR5 and ICOS analysis. Chart shows the percent of SMARTA cells that are CXCR5+ICOS+ Tfh cells. (C) The percent of transferred SMARTA cells that are CXCR5+GL-7+ germinal center Tfh cells. (D) Bcl6 MFI of SMARTA cells. (E) T-bet MFI of SMARTA cells. (F) ICOS MFI on CXCR5− gated effector SMARTA cells 7 days postrechallenge. (G) Chart shows the percent of transferred SMARTA cells that are CXCR5−granzyme Bhi cells 7 days postrechallenge. (H) Chart shows the granzyme B MFI of SMARTA cells in lung 7 days postrechallenge. For (A)–(H), CXCR5−Ly6chi (n = 4) and CXCR5+ (n = 5). Statistically significant p values of <0.05 are indicated and were determined by using a two-tailed unpaired Student’s t test. See also Figure S4. Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions

Figure 5 Tfh Memory CD4+ T Cells Recall a Tfh-like Response Even in B Cell Deficient Recipient Mice CD45.1 congenically marked CXCR5−Ly6chi (blue), CXCR5+Ly6clo (red), and CXCR5+Ly6cint (green) SMARTA memory CD4+ T cell subsets (days 68 and 88 postinfection), and naive SMARTA cells sorted as CD44lo (black) were FACS purified to >97%. We adoptively transferred 8 × 103 purified cells into naive CD45.2+ WT and μMT (B cell-deficient) B6 recipients, and recipient mice were then infected with LCMV Armstrong. The phenotype and function of SMARTA cells were analyzed 7 and 10 days postinfection. (A) Cartoon of experimental setup. (B) Representative CXCR5 and ICOS analyses of CD4+ CD45.1+ gated SMARTA cells 7 and 10 days postinfection. (C) Charts show the frequency of SMARTA cells that are CXCR5+ICOS+ Tfh-like cells in B cell-deficient mice at each time point following infection with LCMV. Data for day 7 were combined from two independent experiments (n = 7 per experimental group) and day 10 were from one experiment (n = 3 per group). Statistically significant p values were determined using a two-tailed unpaired Student’s t test (∗∗∗p < 0.001; ∗∗p < 0.01; ∗p < 0.05). See also Figure S5. Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions

Figure 6 Epigenetic Modifications of the Granzyme B Locus Distinguish Tfh Memory from Th1 Memory CD4+ T Cells (A and B) We transferred 1–2 × 106 CFSE-labeled SMARTA cells into host mice and infected 1 day later. (A) Plots show granzyme B expression and CFSE dilution analysis of SMARTA cells at the indicated time points postinfection. (B) Plot shows granzyme B and T-bet expression analysis of SMARTA cells 2 days postinfection. (C–E) We transferred 2 × 105 SMARTA cells into host mice that were infected 1 day later. (C) Flow cytometry plots show granzyme B and CXCR5 expression of SMARTA cells at the indicated time points postinfection. (D and E) Sorted antigen-specific SMARTA CD4+ Tfh and Th1 cells were isolated from the spleen and purified by FACS at naive, effector, and memory stages of differentiation. Tfh subsets were defined as CXCR5+ Ly6clo, whereas Th1 subsets were defined as CXCR5−Ly6chi. DNA methylation status of (D) Gzmb and (E) Il21, Ifng, and Pdcd1 loci were determined by bisulfite sequencing the genomic DNA from the purified cells. Each horizontal line corresponds to the sequence of an individual clone. Filled circles represent methylated cytosine. Open circles represent nonmethylated cytosine. Representative dot plots from one complete data set are shown. Data are representative of at least three indepentently isolated populations of naive (uninfected CD44lo), effector (days 5–10), and memory (days 49–101) SMARTA cells. See also Figure S6. Immunity 2013 38, 805-817DOI: (10.1016/j.immuni.2013.02.020) Copyright © 2013 Elsevier Inc. Terms and Conditions