Coreceptor Reversal in the Thymus

Slides:



Advertisements
Similar presentations
Volume 34, Issue 3, Pages (March 2011)
Advertisements

Volume 26, Issue 3, Pages (March 2007)
Volume 8, Issue 1, Pages (January 1998)
Marc Hertz, David Nemazee  Immunity 
The Roles of Fas/APO-1 (CD95) and TNF in Antigen-Induced Programmed Cell Death in T Cell Receptor Transgenic Mice  Huey-Kang Sytwu, Roland S Liblau, Hugh.
Volume 38, Issue 6, Pages (June 2013)
Volume 28, Issue 3, Pages (March 2008)
Volume 18, Issue 5, Pages (May 2003)
Volume 28, Issue 1, Pages (January 2008)
Volume 29, Issue 6, Pages (December 2008)
A Negative Regulatory Role for Ig-α during B Cell Development
Volume 21, Issue 2, Pages (August 2004)
Volume 15, Issue 6, Pages (December 2001)
Unexpectedly Complex Regulation of CD4/CD8 Coreceptor Expression Supports a Revised Model for CD4+CD8+ Thymocyte Differentiation  Bruno Lucas, Ronald.
Volume 23, Issue 1, Pages (July 2005)
Fyn Can Partially Substitute for Lck in T Lymphocyte Development
Volume 10, Issue 3, Pages (March 1999)
Acquisition of a Functional T Cell Receptor during T Lymphocyte Development Is Enforced by HEB and E2A Transcription Factors  Mary Elizabeth Jones, Yuan.
Lck Activity Controls CD4/CD8 T Cell Lineage Commitment
A Molecular Map of T Cell Development
Volume 28, Issue 3, Pages (March 2008)
Susan V Outram, Alberto Varas, Carmen V Pepicelli, Tessa Crompton 
Volume 154, Issue 6, Pages (September 2013)
A Two-Step Process for Thymic Regulatory T Cell Development
Volume 36, Issue 3, Pages (March 2012)
Volume 31, Issue 3, Pages (September 2009)
TCR Signal Strength Influences αβ/γδ Lineage Fate
Volume 6, Issue 6, Pages (June 1997)
Volume 22, Issue 3, Pages (March 2005)
Skint-1 Identifies a Common Molecular Mechanism for the Development of Interferon-γ- Secreting versus Interleukin-17-Secreting γδ T Cells  Gleb Turchinovich,
Positive Selection of Dendritic Epidermal γδ T Cell Precursors in the Fetal Thymus Determines Expression of Skin-Homing Receptors  Na Xiong, Chuhlo Kang,
Volume 16, Issue 5, Pages (May 2002)
Identification of a T Lineage-Committed Progenitor in Adult Blood
Abrogation of TGFβ Signaling in T Cells Leads to Spontaneous T Cell Differentiation and Autoimmune Disease  Leonid Gorelik, Richard A Flavell  Immunity 
Volume 22, Issue 2, Pages (February 2005)
Volume 19, Issue 3, Pages (September 2003)
Volume 6, Issue 3, Pages (March 1997)
Volume 8, Issue 5, Pages (May 1998)
Volume 38, Issue 6, Pages (June 2013)
Volume 21, Issue 2, Pages (August 2004)
Volume 15, Issue 4, Pages (October 2001)
Volume 9, Issue 5, Pages (November 1998)
Andrew T Miller, Heather M Wilcox, Zhongbin Lai, Leslie J Berg 
Volume 12, Issue 5, Pages (May 2000)
The Shaping of the T Cell Repertoire
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 24, Issue 1, Pages (January 2006)
Volume 29, Issue 4, Pages (October 2008)
Intrathymic T Cell Development and Selection Proceeds Normally in the Absence of Glucocorticoid Receptor Signaling  Jared F Purton, Richard L Boyd, Timothy.
Volume 23, Issue 1, Pages (July 2005)
Volume 14, Issue 3, Pages (March 2001)
Volume 25, Issue 4, Pages (October 2006)
Volume 31, Issue 6, Pages (December 2009)
Volume 17, Issue 5, Pages (November 2002)
Volume 17, Issue 4, Pages (October 2002)
Volume 27, Issue 5, Pages (November 2007)
Volume 28, Issue 5, Pages (May 2008)
Thomas M. Schmitt, Juan Carlos Zúñiga-Pflücker  Immunity 
Notch 1 Signaling Regulates Peripheral T Cell Activation
Volume 22, Issue 5, Pages (May 2005)
Volume 25, Issue 1, Pages (July 2006)
Volume 31, Issue 6, Pages (December 2009)
TCR Signal Strength Influences αβ/γδ Lineage Fate
Volume 20, Issue 6, Pages (June 2004)
Volume 27, Issue 5, Pages (November 2007)
Volume 23, Issue 4, Pages (October 2005)
Volume 25, Issue 4, Pages (October 2006)
Generation of Functional Thymocytes in the Human Adult
Abrogation of TGFβ Signaling in T Cells Leads to Spontaneous T Cell Differentiation and Autoimmune Disease  Leonid Gorelik, Richard A Flavell  Immunity 
Volume 10, Issue 3, Pages (March 1999)
Presentation transcript:

Coreceptor Reversal in the Thymus Enrico Brugnera, Avinash Bhandoola, Ricardo Cibotti, Qing Yu, Terry I Guinter, Yoshio Yamashita, Susan O Sharrow, Alfred Singer  Immunity  Volume 13, Issue 1, Pages 59-71 (July 2000) DOI: 10.1016/S1074-7613(00)00008-X

Figure 1 Developmental Potential of Newly Arising CD4+8− Thymocytes Generated In Vitro Purified DP thymocytes were obtained from MHCo or ZAPo mice to avoid any possible contamination with preexisting SP T cells. (A) In vitro development of in vitro signaled DP thymocytes. DP thymocytes were stimulated in “signaling cultures” for 12–16 hr either with immobilized anti-TCR+anti-CD2 or with PMA+ionomycin (P+I) and then transferred into nonstimulatory “recovery cultures.” (B) In vivo development of in vitro signaled DP thymocytes. MHCo DP thymocytes were stimulated in signaling cultures for 12–16 hr either with immobilized anti-TCR(H57)+anti-CD2 or with P+I and then directly injected into the thymus of B6 congenic (CD45.1) mice. Two days after injection, host thymi were harvested and subjected to the coreceptor reexpression assay in which thymocytes were stripped of preexisting CD4 and CD8 surface coreceptor molecules by extracellular treatment with pronase and cultured overnight at 37°C, during which the cells reexpress the coreceptor molecules that they are actively synthesizing. Donor thymocytes were assessed for CD4 and CD8 surface expression by four-color flow cytometry in which donor thymocytes were unequivocally identified as CD45.1−CD45.2+ cells. Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)

Figure 2 Phenotypic Evolution of Signaled DP Thymocytes Maintained by IL-7 Purified DP thymocyes were obtained from MHCo or ZAPo mice to avoid possible contamination with SP T cells, stimulated in vitro with P+I, and then transferred into recovery cultures. (A) Effect of signaling on IL-7Rα expression. Surface expression of IL-7Rα chains was assessed on DP thymocytes before and after signaling and recovery cultures. Similar results were obtained by anti-TCR+anti-CD2 stimulation. Shaded curve, negative control antibody; long dash, starting DP thymocytes; short dash, DP thymocytes after signaling culture; solid line, DP thymocytes after recovery culture. (B) Phenotypic changes in signaled DP thymocytes maintained in culture with IL-7. ZAPo DP thymocytes were stimulated with P+I and then transferred to recovery cultures containing recombinant murine IL-7. The numbers under each profile indicate the number of viable cells present, and the numbers in each box indicate the percentage of total cells within that box. In the absence of either Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)

Figure 3 Molecular Basis for Coreceptor Reversal in Newly Generated CD4+8− Thymocytes (A) Reversal of coreceptor protein and RNA expression in newly arising CD4+8− thymocytes. DP thymocytes were obtained from ZAPoMHCIIo double-deficient mice to avoid the possibility of contamination with SP T cells and were signaled in vitro either with P+I (1a) or medium alone (1b). At the conclusion of signaling culture, stimulated cells were pronased to remove preexisting surface coreceptor molecules and then placed in recovery cultures during which the cells reexpressed coreceptor molecules that they were actively synthesizing (2a). Cells that reappeared as CD4+8− after pronase treatment were electronically sorted (2b) and cultured with recombinant murine IL-7 (3b). Remarkably, purified CD4+8− thymocytes reexpressed CD8 surface protein after 1 day in IL-7. Such cells were then subjected to the coreceptor reexpression assay to identify the coreceptor molecules that they were actively synthesizing (4a and 4b). Semiquantitative RT–PCR for HPRT, CD4, and CD8 mRNA transcripts were performed on the indicated cell populations. (B) Reversal of coreceptor gene transcription in newly arising CD4+8− thymocytes. Nuclear run-on assays were performed on the indicated cell populations to assess CD4, CD8α, and HPRT transcription. Relative CD4 and CD8α transcription was quantitated by phosphorimaging and normalized to HPRT as a positive control and to Bluescript as a negative control. CD4 and CD8α gene transcription in medium-stimulated DP thymocytes were set equal to 1.0. Relative CD4 and CD8α gene transcription is displayed both as the original phosphorimage and as a bar graph. (C) CD4 silencer requirement for termination of CD4 promoter activity in newly arising CD4+8− thymocytes. DP thymocytes were obtained from CD4 promoter transgenic mice whose expression of the hCD2 transgene is controlled by CD4 regulatory elements that either include the CD4 silencer (line 30) or lack the CD4 silencer (line 7200). DP thymocytes were signaled by P+I, cultured with IL-7, and then pronased stripped to identify the coreceptor molecules they were synthesizing. Note that pronase treatment also strips preexisting hCD2 molecules from the surface of these cells (data not shown), so that reexpression of CD4, CD8, and hCD2 after pronase treatment reflects transport to the surface of newly synthesized proteins. Shaded curve, negative control antibody staining. Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)

Figure 4 Developmental Potential of Intermediate Thymocytes Generated In Vivo (A) IL-7Rα expression on subpopulations of CD4+8lo thymocytes. CD4+8lo thymocytes were obtained from B6 mice by electronic cell sorting, pronase treated, and cultured overnight to permit the cells to reexpress the coreceptor molecules they were actively synthesizing. CD4+8lo thymocytes from normal mice contain cells actively synthesizing both CD4 and CD8 coreceptors and so reappear after pronase treatment as CD4+8+; cells actively synthesizing only CD4 coreceptors and so reappear after pronase treatement as CD4+8− cells; and cells actively synthesizing only CD8 coreceptors and so reappear after pronase treatment as CD4−8+ cells. As pronase treatment also strips preexisting IL-7Rα chains from the cell surface, we assessed IL-7Rα synthesis and reexpression in each subpopulation after pronase treatment. (B) Coreceptor reversal in intermediate thymocytes generated in vivo. Mature CD4+8− and immature CD4+8lo thymocytes were obtained from β2mo mice by electronic sorting. The sorted cells were then pronase treated and placed into culture with medium for 1 day to permit them to reexpress the coreceptor molecules they were actively synthesizing. After pronase treatment, cells that reappeared either as CD4+8− or CD4+8+ were purified by another electronic sort and placed in overnight culture with IL-7. The cultured cells were then assessed for surface CD4 and CD8 expression. Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)

Figure 5 Inhibition of Coreceptor Reversal in Intermediate Thymocytes by TCR Signaling (A) Effect of TCR stimulation on intermediate thymocytes generated in vivo. Intermediate thymocytes that had been generated in vivo were obtained by electronic sorting of CD4+8lo cells from β2mo mice, pronase stripped, and cultured in medium to verify that the sorted cells were devoid of CD4−8+ synthesizing thymocytes (1a). Alternatively, the sorted cells were pronase stripped the following day and cultured overnight with IL-7 either with or without concurrent TCR stimulation (2a and 2b). The appearance of CD4−8+ thymocytes was blocked by TCR stimulation despite the presence of IL-7 (2a and 2b). (B) Effect of P+I restimulation on coreceptor reversal in in vitro generated CD4+8− thymocytes. Intermediate thymocytes were generated in vitro by P+I stimulation of MHCo DP thymocytes and were purified by electronically sorting cells that reappeared after pronase treatment as CD4+8− (2a and 2b). These newly arising CD4+8− cells were then cultured in IL-7 with or without P+I restimulation (3a and 3b). The following day the cultured cells were treated with pronase to identify coreceptor synthesis in these cells (4a and 4b). Note that the appearance of CD4−8+ thymocytes was blocked by TCR signals generated by P+I restimulation despite the presence of IL-7 (4a and 4b). Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)

Figure 6 Developmental Potential of Intermediate Thymocytes and Cytokine Dependence of CD8 T Cell Differentiation in the Thymus (A) In vivo precursor/progeny relationship between intermediate thymocytes and CD8SP T cells. To obtain intermediate thymocytes for intrathymic transfer with a minimal amount of in vitro manipulation, β2mo thymocytes that lack CD4+8− cells were pronase treated and cultured so that both mature CD4SP T cells and intermediate CD4+8− thymocytes would reappear as CD4+8− after pronase treatment (1a). In contrast, in the absence of pronase treatment, only mature CD4SP T cells appear as CD4+8− cells (1c). Each CD4+8− cell population was purified by electronic sorting (1b and 1d) and transferred into the thymus of a host CD45.1 mouse. Three days after intrathymic transfer, CD4 and CD8 expression on donor derived thymocytes was assessed by four-color flow cytometry, with donor-derived cells unequivocally identified as CD45.1-CD45.2+ cells (4a and 4b). (B) Intrathymic blockade of early CD8 T cell generation by antibodies to both components of surface IL-7 receptors. B6 fetal thymocytes (day 17.5) were placed into fetal thymic organ cultures with medium, a mix of rat antibodies to the α and γc chains of the IL-7R, or control rat IgG. On day 3, thymocytes were harvested, pronase stripped, and cultured overnight to reveal the coreceptor molecules they were actively synthesizing. The pronased cells were assessed by three-color flow cytometry for CD4, CD8, and TCRβ expression. In the one-color TCRβ histograms of pronased-stripped fetal thymocytes, the dashed line represents DP cells, the solid line represents CD4+8− cells, and the shaded curve represents CD4−8+ cells. Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)

Figure 7 Kinetic Signaling Model Of Lineage Determination An idealized flow cytometry profile of developing thymocytes is presented in the top panel. TCR-signaled DP thymocytes initially terminate CD8 transcription and upregulate IL-7Rα expression to become intermediate CD4+8− thymocytes that appear phenotypically as CD4+8lo cells. If TCR signaling persists, intermediate thymocytes differentiate into CD4SP T cells, because persistent TCR signals block IL-7-induced coreceptor reversal. However, if TCR signaling ceases, intermediate thymocytes undergo cytokine-dependent “coreceptor reversal” and differentiate into CD8SP T cells. The middle and bottom panels are pictorial representations by the kinetic signaling model of why CD8-dependent (MHC I) TCR signals drive development of CD8SP T cells, whereas CD8-independent (MHC II) TCR signals drive development of CD4SP T cells. In the middle panel, coengagement of MHC I/peptide complexes by TCR and CD8 surface molecules on DP thymocytes generates TCR signals that initially terminate CD8 transcription. CD8-dependent TCR signaling ceases when CD8 surface protein expression declines, allowing the cells to undergo coreceptor reversal and to differentiate into CD8SP T cells. In the bottom panel, engagement of MHC II/peptide complexes by TCR and CD4 surface molecules on DP thymocytes generates TCR signals that also terminate CD8 transcription. However, CD8-independent TCR signaling persists despite the decline in CD8 surface protein expression, blocking coreceptor reversal and leading to differentiation into CD4SP T cells. Immunity 2000 13, 59-71DOI: (10.1016/S1074-7613(00)00008-X)