Expression profiles of adhesion molecules on naïve T cells in bone marrow grafts of healthy donors treated with granulocyte colony-stimulating factor

2009 ◽  
Vol 21 (4) ◽  
pp. 228-233 ◽  
Author(s):  
Ying-Jun Chang ◽  
Xiang-Yu Zhao ◽  
Ming-Rui Huo ◽  
Xiao-Jun Huang
Blood ◽  
2000 ◽  
Vol 95 (8) ◽  
pp. 2484-2490 ◽  
Author(s):  
Mario Arpinati ◽  
Cherie L. Green ◽  
Shelly Heimfeld ◽  
Jill E. Heuser ◽  
Claudio Anasetti

Peripheral blood stem cells (PBSC) obtained from granulocyte-colony stimulating factor (G-CSF)-mobilized donors are increasingly used for allogeneic transplantation. Despite a 10-fold higher dose of transplanted T cells, acute graft-versus-host disease (GVHD) does not develop in higher proportion in recipients of PBSC than in recipients of marrow. T cells from G-CSF-treated experimental animals preferentially produce IL-4 and IL-10, cytokines characteristic of Th2 responses, which are associated with diminished GVHD-inducing ability. We hypothesized that G-CSF-mobilized PBSC contain antigen-presenting cells, which prime T-lymphocytes to produce Th2 cytokines. Two distinct lineages of dendritic cells (DC) have been described in humans, DC1 and DC2, according to their ability to induce naive T-cell differentiation to Th1 and Th2 effector cells, respectively. We have used multicolor microfluorometry to enumerate DC1 and DC2 in the peripheral blood of normal donors. G-CSF treatment with 10 to 16 μg/kg per day for 5 days increased peripheral blood DC2 counts from a median of 4.9 × 106/L to 24.8 × 106/L (P = .0009), whereas DC1 counts did not change. Purified DC1, from either untreated or G-CSF treated donors, induced the proliferation of allogeneic naive T cells, but fresh DC2 were poor stimulators. Tumor necrosis factor- (TNF-)-activated DC1 induced allogeneic naive T cells to produce IFN-γ, which is typical of Th1 responses, whereas TNF--activated DC2 induced allogeneic naive T cells to produce IL-4 and IL-10, which are typical of Th2 responses. PBSC transplants contained higher doses of DC2 than marrow transplants (median, 2.4 × 106/kg versus 0.5 × 106/kg) (P = .006), whereas the dose of DC1 was comparable. Thus, it is conceivable that transplantation of G-CSF-stimulated PBSC does not result in overwhelming acute GVHD because the graft contains predominantly Th2-inducing DC. Adoptive transfer of purified DC2 may be exploited to induce immune deviation after transplantation of hematopoietic stem cells or organ allografts.


Blood ◽  
2000 ◽  
Vol 95 (8) ◽  
pp. 2484-2490 ◽  
Author(s):  
Mario Arpinati ◽  
Cherie L. Green ◽  
Shelly Heimfeld ◽  
Jill E. Heuser ◽  
Claudio Anasetti

Abstract Peripheral blood stem cells (PBSC) obtained from granulocyte-colony stimulating factor (G-CSF)-mobilized donors are increasingly used for allogeneic transplantation. Despite a 10-fold higher dose of transplanted T cells, acute graft-versus-host disease (GVHD) does not develop in higher proportion in recipients of PBSC than in recipients of marrow. T cells from G-CSF-treated experimental animals preferentially produce IL-4 and IL-10, cytokines characteristic of Th2 responses, which are associated with diminished GVHD-inducing ability. We hypothesized that G-CSF-mobilized PBSC contain antigen-presenting cells, which prime T-lymphocytes to produce Th2 cytokines. Two distinct lineages of dendritic cells (DC) have been described in humans, DC1 and DC2, according to their ability to induce naive T-cell differentiation to Th1 and Th2 effector cells, respectively. We have used multicolor microfluorometry to enumerate DC1 and DC2 in the peripheral blood of normal donors. G-CSF treatment with 10 to 16 μg/kg per day for 5 days increased peripheral blood DC2 counts from a median of 4.9 × 106/L to 24.8 × 106/L (P = .0009), whereas DC1 counts did not change. Purified DC1, from either untreated or G-CSF treated donors, induced the proliferation of allogeneic naive T cells, but fresh DC2 were poor stimulators. Tumor necrosis factor- (TNF-)-activated DC1 induced allogeneic naive T cells to produce IFN-γ, which is typical of Th1 responses, whereas TNF--activated DC2 induced allogeneic naive T cells to produce IL-4 and IL-10, which are typical of Th2 responses. PBSC transplants contained higher doses of DC2 than marrow transplants (median, 2.4 × 106/kg versus 0.5 × 106/kg) (P = .006), whereas the dose of DC1 was comparable. Thus, it is conceivable that transplantation of G-CSF-stimulated PBSC does not result in overwhelming acute GVHD because the graft contains predominantly Th2-inducing DC. Adoptive transfer of purified DC2 may be exploited to induce immune deviation after transplantation of hematopoietic stem cells or organ allografts.


Blood ◽  
1995 ◽  
Vol 86 (12) ◽  
pp. 4422-4429 ◽  
Author(s):  
L Pan ◽  
J Jr Delmonte ◽  
CK Jalonen ◽  
JL Ferrara

The incidence and severity of acute graft-versus-host disease (GVHD) after allogeneic transplantation using peripheral blood progenitor cells mobilized by granulocyte colony-stimulating factor (G-CSF) appear to be no worse than those after bone marrow transplantation, despite the presence of large numbers of T cells in the donor infusion. Experimental studies have shown that type-1 T cells (secreting interleukin-2 [IL-2] and interferon-gamma) mediate acute GVHD, whereas type-2 T cells (secreting IL-4 and IL-10) can prevent acute GVHD. We tested the hypothesis that G-CSF modulates T-cell function toward a type-2 response and thus reduces the severity of acute GVHD. B6 mice were injected with G-CSF or diluent for 4 days, and their splenic T cells were stimulated in vitro with alloantigen or mitogen in the absence of G-CSF. T cells from G-CSF-treated mice showed a significant increase in IL-4 production, with a simultaneous decrease in IL-2 and interferon-gamma production in response to both stimuli. We also examined the effect of G-CSF pretreatment of donors in a GVHD model (B6- ->B6D2F1). Survival was significantly improved in recipients of G-CSF- treated donors. Concanavalin-A-induced cytokine production at day 13 after transplantation also showed an increase in IL-4 along with a decrease in IL-2 and IFN-gamma production by splenocytes from recipients of G-CSF-treated bone marrow and T cells. These data show that pretreatment of donors with G-CSF polarizes donor T cells toward the production of type-2 cytokines, which is associated with reduced type-1 cytokine production and reduced severity of acute GVHD.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3147-3147
Author(s):  
Matthew J. Olnes ◽  
Susan Leitman ◽  
Angelique Biancotto ◽  
J. Philip McCoy ◽  
Susan Miranda ◽  
...  

Abstract Abstract 3147 Poster Board III-84 Recent reports of chromosomal and immunological abnormalities in healthy donors receiving granulocyte colony stimulating factor (G-CSF) have raised concerns among hematologists that this popular cytokine may promote genomic instability or alter immune surveillance (Pampilon D et al Transfusion 2008; 48(7):1495-501). We previously reported that G-CSF altered the Th1:Th2 ratio in healthy individuals following short-term administration (Blood 2000 Apr 1;95(7):2269-74), but reported no new karyotypic abnormities after in vitro culture of bone marrow mononuclear cells with pharmacological doses of G-CSF (Proc Natl Acad Sci 2006 Sep 26;103(39):14483-8). There is no systematic study of the long-term effects of administering granulocyte colony stimulating factor (G-CSF) to healthy individuals. We examined CD34 cells of 10 healthy stem cell donors after they had received 10 mcg/kg G-CSF for 4 days; fluorescent in situ hybridization (FISH) was the method employed to monitor chromosomal changes. We also studied 37 healthy granulocyte donors who received G-CSF (5ug/Kg x 1 day) and dexamethosone for up to 42 times (median= 15; range 6-42) using FISH to examine chromosomes 7 and 8 and flow cytometry to define their T cell subsets. FISH did not detect chromosomal abnormalities in the CD34 cells of 10 donors mobilized with G-CSF; neither could monosomy 7 cells be isolated after culturing cells in media with 400 ng/mL G-CSF (previously shown to support outgrowth of monosomy 7 cells) for two weeks. Furthermore, FISH did not detect aneuploidy in the 37 regular granulocyte donors. Evaluation of T cell subsets by flow cytometry demonstrated similar percentages of CD4+ T cells in 18 granulocyte donors as compared to 23 untreated controls (57.5% vs 56.5%). However, donors had increased numbers of CD4+TNFαa+ Th1 T cells and decreased CD4+IL-6+ Th2 T cells (4.2% vs 1.6%, P= 0.0003 and 11% vs. 35%, P=0.04 respectively), while the donor Th2 subset expressed significantly more IL-6 per cell (P<0.01). CD4+CD25+FoxP3+ regulatory T cells (Tregs) were significantly increased in G-CSF-treated donors (10.1% vs 6.0%, P<0.0001), while Th17 T cells were not significantly different (2.4% vs 0.7%, P=0.423). G-CSF does not produce chromosomal abnormalities of monosomy 7 or trisomy 8 in healthy SCT donors or in serially treated granulocyte donors. However, there are significant changes in T cell subsets that modulate the immune response. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3051-3051
Author(s):  
Hui Liu ◽  
Xuan Du ◽  
Yinkui Chen ◽  
Li Xuan ◽  
Xiuli Wu ◽  
...  

Abstract Abstract 3051 Background Peripheral blood stem cells (PBSCs) obtained from granulocyte colony-stimulating factor (G-CSF)-mobilized donors have been used more frequently than bone marrow stem cells as the source of cells in allogeneic hematopoietic stem cell transplantation (allo-HSCT). Although G-CSF–mobilized PBSC grafts contain more mature T cells than bone marrow cell grafts, the incidence and severity of graft-versus-host disease (GVHD) were not elevated. G-CSF-induced immune tolerance might be via inducing T helper type 2 (Th2) polarization, the promotion of regulatory T cell and tolerogenic dendritic cell (DC) differentiation. However, these mechanisms are not fully understood. Human leucocyte antigen-G (HLA-G) is a tolerogenic molecule which participates in the regulation of immune response. In this study, to explore the mechanisms of G-CSF-induced immunoregulation, the effect of G-CSF on blood cells expressing HLA-G was studied. Methods Membrane-bound HLA-G (mHLA-G) was detected using flow cytometry analysis; soluble HLA-G (sHLA-G), interferon gamma (IFN-γ) and interleukin 10 (IL-10) were determined by enzyme-linked immunosorbent assay (ELISA); HLA-Gpos cells were isolated using flow cytometry, and mixed leukocytes reaction (MLR) was carried out to assess the suppressive effect of HLA-Gpos cells. Results CD3+CD4+ T cells, CD3+CD8+ T cells, CD19+ cells and CD14+ cells all expressed mHLA-G in peripheral blood (PB) and bone marrow (BM) from 18 healthy donors before and after G-CSF mobilization. The levels of sHLA-G and mHLA-G on these cells in PB and BM all increased significantly after G-CSF mobilization (all P<0.05). The levels of sHLA-G and mHLA-G on these cells in BM were all higher than that in PB, including before and after G-CSF mobilization (all P<0.05). Bone marrow mononuclear cells (BMMCs) were stimulated with G-CSF in vitro, and the levels of mHLA-G on these cells and sHLA-G in culture supernatant all increased significantly after BMMCs cultivated with G-CSF for 24 hours (all P<0.001). In addition, the levels of IFN-γ and IL-10 also elevated in culture supernatant (both P<0.05). Antibodies blocking experiments for IL-10 and IFN-γ showed that IL-10 and IFN-γ were not necessary for G-CSF-induced HLA-G expression of blood cells. The results of MLR showed that HLA-Gpos cells were able to suppress the proliferation of allogeneic lymphocytes. Conclusion G-CSF could directly induce blood cells expressing HLA-G, which might be another mechanism of G-CSF-mediated immunoregulation in G-CSF–mobilized PBSC transplants. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document