by Sheng F. Cai, Xuefang Cao, Anjum Hassan, Todd A

Slides:



Advertisements
Similar presentations
Recombinant CD95-Fc (APG101) prevents graft-versus-host disease in mice without disabling antitumor cytotoxicity and T-cell functions by Natalie Hartmann,
Advertisements

Joseph H. Chewning, Weiwei Zhang, David A. Randolph, C
Reduced Graft-versus-Host Disease in C3-Deficient Mice Is Associated with Decreased Donor Th1/Th17 Differentiation  Qing Ma, Dan Li, Roza Nurieva, Rebecca.
Unique patterns of surface receptors, cytokine secretion, and immune functions distinguish T cells in the bone marrow from those in the periphery: impact.
Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease by Tangsheng Yi, Ying Chen, Lin Wang,
Host-Derived CD8+ Dendritic Cells Protect Against Acute Graft-versus-Host Disease after Experimental Allogeneic Bone Marrow Transplantation  Michael Weber,
The Fifth Epidermal Growth Factor–like Region of Thrombomodulin Alleviates Murine Graft-versus-Host Disease in a G-Protein Coupled Receptor 15 Dependent.
Influence of Donor Microbiota on the Severity of Experimental Graft-versus-Host- Disease  Isao Tawara, Chen Liu, Hiroya Tamaki, Tomomi Toubai, Yaping Sun,
Stromal-Derived Factor-1α and Interleukin-7 Treatment Improves Homeostatic Proliferation of Naïve CD4+ T Cells after Allogeneic Stem Cell Transplantation 
Host-Derived Interleukin-18 Differentially Impacts Regulatory and Conventional T Cell Expansion During Acute Graft-Versus-Host Disease  Robert Zeiser,
Extracorporeal Photopheresis Attenuates Murine Graft-versus-Host Disease via Bone Marrow–Derived Interleukin-10 and Preserves Responses to Dendritic Cell.
IL-2–Targeted Therapy Ameliorates the Severity of Graft-versus-Host Disease: Ex Vivo Selective Depletion of Host-Reactive T Cells and In Vivo Therapy 
Host conditioning with total lymphoid irradiation and antithymocyte globulin prevents graft-versus-host disease: the role of CD1-reactive natural killer.
Adoptive transfer of allogeneic tumor-specific T cells mediates effective regression of large tumors across major histocompatibility barriers by Andrea.
Identification of Stem Cell Transcriptional Programs Normally Expressed in Embryonic and Neural Stem Cells in Alloreactive CD8+ T Cells Mediating Graft-versus-Host.
Juyang Kim, Wongyoung Kim, Hyun J. Kim, Sohye Park, Hyun-A
CCR2 is required for CD8-induced graft-versus-host disease
Notch signaling is a critical regulator of allogeneic CD4+ T-cell responses mediating graft-versus-host disease by Yi Zhang, Ashley R. Sandy, Jina Wang,
by Yoshinobu Maeda, Pavan Reddy, Kathleen P
by Dennis Adeegbe, Robert B. Levy, and Thomas R. Malek
Ping Zhang, Jieying Wu, Divino Deoliveira, Nelson J. Chao, Benny J
Apoptotic Donor Leukocytes Limit Mixed-Chimerism Induced by CD40-CD154 Blockade in Allogeneic Bone Marrow Transplantation  Jian-ming Li, John Gorechlad,
Human NK cell development in NOD/SCID mice receiving grafts of cord blood CD34+ cells by Christian P. Kalberer, Uwe Siegler, and Aleksandra Wodnar-Filipowicz.
by Norman Nausch, Ioanna E
Ikaros-Notch axis in host hematopoietic cells regulates experimental graft-versus-host disease by Tomomi Toubai, Yaping Sun, Isao Tawara, Ann Friedman,
Macrophages from C3-deficient mice have impaired potency to stimulate alloreactive T cells by Wuding Zhou, Hetal Patel, Ke Li, Qi Peng, Marie-Bernadette.
Ex Vivo Rapamycin Generates Th1/Tc1 or Th2/Tc2 Effector T Cells With Enhanced In Vivo Function and Differential Sensitivity to Post-transplant Rapamycin.
IL-21 blockade reduces graft-versus-host disease mortality by supporting inducible T regulatory cell generation by Christoph Bucher, Lisa Koch, Christine.
B7-H3 expression in donor T cells and host cells negatively regulates acute graft-versus-host disease lethality by Rachelle G. Veenstra, Ryan Flynn, Katharina.
LBH589 Enhances T Cell Activation In Vivo and Accelerates Graft-versus-Host Disease in Mice  Dapeng Wang, Cristina Iclozan, Chen Liu, Changqing Xia, Claudio.
IL-12hi Rapamycin-Conditioned Dendritic Cells Mediate IFN-γ–Dependent Apoptosis of Alloreactive CD4+ T Cells In Vitro and Reduce Lethal Graft-Versus-Host.
Expansion of FOXP3high regulatory T cells by human dendritic cells (DCs) in vitro and after injection of cytokine-matured DCs in myeloma patients by Devi.
by Éric Aubin, Réal Lemieux, and Renée Bazin
The histone methyltransferase Ezh2 is a crucial epigenetic regulator of allogeneic T-cell responses mediating graft-versus-host disease by Shan He, Fang.
Immune Tolerance to Self-Major Histocompatability Complex Class II Antigens after Bone Marrow Transplantation: Role of Regulatory T Cells  Allan D. Hess,
Simple conditioning with monospecific CD4+CD25+ regulatory T cells for bone marrow engraftment and tolerance to multiple gene products by David-Alexandre.
Blocking LFA-1 Activation with Lovastatin Prevents Graft-versus-Host Disease in Mouse Bone Marrow Transplantation  Yang Wang, Dan Li, Dan Jones, Roland.
IFNγ differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract by Angela C. Burman, Tatjana Banovic,
IL-6 Blockade Attenuates the Development of Murine Sclerodermatous Chronic Graft- Versus-Host Disease  Doanh Le Huu, Takashi Matsushita, Guihua Jin, Yasuhito.
IL-21 inhibits T cell IL-2 production and impairs Treg homeostasis
IL-17 Gene Ablation Does Not Impact Treg-Mediated Suppression of Graft-Versus-Host Disease after Bone Marrow Transplantation  Lucrezia Colonna, Mareike.
Evelyn C. Nieves, Tomomi Toubai, Daniel C
Pharmacologic Expansion of Donor-Derived, Naturally Occurring CD4+Foxp3+ Regulatory T Cells Reduces Acute Graft-versus-Host Disease Lethality Without.
by Anil Dangi, Lei Zhang, Xiaomin Zhang, and Xunrong Luo
Absence of donor Th17 leads to augmented Th1 differentiation and exacerbated acute graft-versus-host disease by Tangsheng Yi, Dongchang Zhao, Chia-Lei.
An Essential Role for IFN-γ in Regulation of Alloreactive CD8 T Cells Following Allogeneic Hematopoietic Cell Transplantation  Wannee Asavaroengchai,
Therapeutic Benefit of Bortezomib on Acute Graft-versus-Host Disease Is Tissue Specific and Is Associated with Interleukin-6 Levels  Chien-Chun Steven.
Blocking Activator Protein 1 Activity in Donor Cells Reduces Severity of Acute Graft- Versus-Host Disease through Reciprocal Regulation of IL-17–Producing.
Essential Role of Interleukin-12/23p40 in the Development of Graft-versus-Host Disease in Mice  Yongxia Wu, David Bastian, Steven Schutt, Hung Nguyen,
T helper17 Cells Are Sufficient But Not Necessary to Induce Acute Graft-Versus-Host Disease  Cristina Iclozan, Yu Yu, Chen Liu, Yaming Liang, Tangsheng.
The Triterpenoid CDDO-Me Delays Murine Acute Graft-versus-Host Disease with the Preservation of Graft-versus-Tumor Effects after Allogeneic Bone Marrow.
Host Basophils Are Dispensable for Induction of Donor T Helper 2 Cell Differentiation and Severity of Experimental Graft-versus-Host Disease  Isao Tawara,
A Radio-Resistant Perforin-Expressing Lymphoid Population Controls Allogeneic T Cell Engraftment, Activation, and Onset of Graft-versus-Host Disease in.
Dynamic Change and Impact of Myeloid-Derived Suppressor Cells in Allogeneic Bone Marrow Transplantation in Mice  Dapeng Wang, Yu Yu, Kelley Haarberg,
Mammalian Target of Rapamycin Inhibitors Permit Regulatory T Cell Reconstitution and Inhibit Experimental Chronic Graft-versus-Host Disease  Haruko Sugiyama,
Volume 10, Issue 5, Pages (May 1999)
Tracking ex vivo-expanded CD4+CD25+ and CD8+CD25+ regulatory T cells after infusion to prevent donor lymphocyte infusion-induced lethal acute graft-versus-host.
Volume 27, Issue 4, Pages (October 2007)
B7.2−/− Mature Dendritic Cells Generate T-Helper 2 and Regulatory T Donor Cells in Fetal Mice after In Utero Allogeneic Bone Marrow Transplantation  Swati.
Volume 27, Issue 3, Pages (September 2007)
CD25 expression distinguishes functionally distinct alloreactive CD4+ CD134+ (OX40+) T-cell subsets in acute graft-versus-host disease  Philip R Streeter,
Brile Chung, Eric Dudl, Akira Toyama, Lora Barsky, Kenneth I. Weinberg 
Post-Transplantation Cyclophosphamide and Ixazomib Combination Rescues Mice Subjected to Experimental Graft-versus-Host Disease and Is Superior to Either.
Raimon Duran-Struuck, Isao Tawara, Kathi Lowler, Shawn G
IAPs protect host target tissues from graft-versus-host disease in mice by Tomomi Toubai, Corinne Rossi, Katherine Oravecz-Wilson, Chen Liu, Cynthia Zajac,
Roles of CD28, CTLA4, and Inducible Costimulator in Acute Graft-versus-Host Disease in Mice  Jun Li, Kenrick Semple, Woong-Kyung Suh, Chen Liu, Fangping.
Volume 26, Issue 6, Pages (June 2007)
Rapamycin inhibits IL-4—induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo by Holger Hackstein, Timucin Taner,
Volume 37, Issue 2, Pages (August 2012)
Volume 10, Issue 5, Pages (February 2015)
Presentation transcript:

Granzyme B is not required for regulatory T cell–mediated suppression of graft-versus-host disease by Sheng F. Cai, Xuefang Cao, Anjum Hassan, Todd A. Fehniger, and Timothy J. Ley Blood Volume 115(9):1669-1677 March 4, 2010 ©2010 by American Society of Hematology

Alloactivated Treg cells express granzyme B during in vitro MLRs Alloactivated Treg cells express granzyme B during in vitro MLRs. The 129/SvJ WT (H-2Kb) splenocytes were cultured with irradiated (2000 cGy) Balb/c WT (H-2Kd) splenocytes in complete medium supplemented with 50 U/mL IL-2. Alloactivated Treg cells express granzyme B during in vitro MLRs. The 129/SvJ WT (H-2Kb) splenocytes were cultured with irradiated (2000 cGy) Balb/c WT (H-2Kd) splenocytes in complete medium supplemented with 50 U/mL IL-2. At various time points, splenocytes were harvested and analyzed by flow cytometry for granzyme B expression. (A) Representative flow plots of granzyme B expression, gated on naive and day 6 MLR-stimulated CD8+ T cells. (B) Representative flow plots of granzyme B expression, gated on naive and day 6 MLR-stimulated CD4+Foxp3+ Treg cells. (C) Summary graph of percentage granzyme B–expressing Treg cells (n = 3 independent MLR cultures per time point). Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology

Alloactivated Treg cells express granzyme B in a mouse model of GVHD in vivo. Alloactivated Treg cells express granzyme B in a mouse model of GVHD in vivo. Lethally irradiated Balb/c mice were reconstituted with 2 × 106 bone marrow cells and 2 × 106 T cells derived from 129/SvJ WT mice. Splenocytes were harvested from recipient mice on days 3 to 6 for flow cytometric analysis of granzyme B expression in donor-derived Treg cells. (A) Representative flow plots, gated on H-2Kb+Foxp3+ Treg cells, are shown. (B) Summary graph of percentage granzyme B–expressing Treg cells is shown (n = 3 mice per time point). Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology

Granzyme B is not required for Treg cell–mediated suppression of ConA-activated and alloactivated Teff-cell proliferation in vitro. Granzyme B is not required for Treg cell–mediated suppression of ConA-activated and alloactivated Teff-cell proliferation in vitro. (A) Suppression of CD4+CD25− Teff-cell proliferation stimulated by ConA in the presence of syngeneic T cell–depleted, irradiated APCs. (B) Representative flow plot of granzyme B expression, gated on wild-type Treg cells cultured for 3 days with ConA-activated Teff cells under maximal suppression conditions (1:1). (C) Dose-dependent suppression of 129/SvJ CD4+CD25− Teff-cell proliferation stimulated by fully mismatched Balb/c T cell–depleted APCs. (D) Representative flow plot of granzyme B expression, gated on wild-type Treg cells cultured for 5 days with alloactivated Teff cells under maximal suppression conditions (1:1). All data shown are representative of 3 independent experiments. Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology

In vivo alloactivated Treg cells do not require granzyme B to suppress Teff-cell proliferation ex vivo. In vivo alloactivated Treg cells do not require granzyme B to suppress Teff-cell proliferation ex vivo. (A) Experimental protocol for generation of WT and Gzmb−/− GVHD-activated Treg cells: 2 × 106 T cells from 129/SvJ WT or Gzmb−/− FIG reporter mice were injected intravenously in lethally irradiated (900 cGy) Balb/c hosts, together with 2 × 106 bone marrow cells from WT (non-FIG) mice. Four days after transplantation, splenic WT or Gzmb−/− CD4+GFP+ Treg cells were sort-purified and cultured ex vivo with DDAO-SE–stained CD4+CD25− Teff cells for 3 days. T cells were either unstimulated or stimulated with CD3/CD28 beads. (B) Representative flow plot and histogram of Teff-cell proliferation (ie, loss of DDAO-SE staining) in the presence or absence of CD3/CD28 beads. (C) Dose-dependent inhibition of Teff-cell proliferation mediated by wild-type or Gzmb−/− GVHD-activated Treg cells. (D) Summary graph of normalized data from 3 independent experiments. Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology

Treg cells do not require granzyme B to rescue hosts from GVHD lethality or to prevent GVHD target organ damage. Treg cells do not require granzyme B to rescue hosts from GVHD lethality or to prevent GVHD target organ damage. Lethally irradiated (900 cGy) Balb/c mice received 2 × 106 129/SvJ TCD BM cells with or without 4 × 105 129/SvJ CD25− Teff cells (both CD4+ and CD8+) and either (A) 4 × 105 or (B) 2 × 105 wild-type or Gzmb−/− CD4+CD25+ Treg cells. Kaplan-Meier survival curves of recipient mice, pooled from 2 independent experiments (n = 10 mice per group), are shown. (C) Seven days after transplantation, 3 mice per experimental group (as outlined in panel A) were killed, and portions of lung, liver, and gut were prepared for histopathologic analysis. There was no statistically significant difference between groups receiving WT or Gzmb−/− Treg cells. Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology

Treg cells do not require granzyme B to suppress production of GVHD-associated cytokines in vivo. Treg cells do not require granzyme B to suppress production of GVHD-associated cytokines in vivo. Lethally irradiated (900 cGy) Balb/c mice received 2 × 106 129/SvJ TCD BM cells with or without 4 × 105 129/SvJ CD25− Teff cells (both CD4+ and CD8+) and 4 × 105 wild-type or Gzmb−/− CD4+CD25+ Treg cells. Seven days after transplantation, serum was harvested and analyzed via cytometric bead array for the production of (A) IL-2, (B) IL-4, (C) IL-5, (D) IL-10, (E) granulocyte-macrophage colony-stimulating factor, and (F) interferon-γ. There was no statistically significant difference between groups receiving WT or Gzmb−/− Treg cells. Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology

Model-dependent role of granzyme B in Treg cell–mediated suppressive function. Model-dependent role of granzyme B in Treg cell–mediated suppressive function. Schematic illustrating the differential phenotypes observed with Gzmb−/− Treg cells in suppressing antitumor responses and GVHD. Sheng F. Cai et al. Blood 2010;115:1669-1677 ©2010 by American Society of Hematology