Granulocyte Colony-Stimulating Factor Mobilization Affects The Expression Of Regulatory γδ T Cells

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 902-902
Author(s):  
Li Xuan ◽  
Xiuli Wu ◽  
Sijian Yu ◽  
Zhengshan Yi ◽  
Yu Zhang ◽  
...  

Abstract Background The immune modulatory effect of granulocyte colony-stimulating factor (G-CSF) on T cells resulted in an unexpected low incidence of graft-versus-host disease (GVHD) in allogeneic peripheral blood stem cell transplantation. Our previous studies demonstrated that G-CSF mobilization influenced the distribution and clonality of TRGV and TRDV repertoire (T cell receptors of γδ T cells), and significant positive correlation was observed between the invariable clonality of TRDV1 gene repertoire after G-CSF mobilization and low incidence of GVHD in recipients (P=0.015, OR=0.047) (Li Xuan et al. Journal of Translational Medicine 2011). Regulatory γδ T cells (γδ Tregs), which express Foxp3 and primarily belong to CD27+CD25high phenotype, are a novel subset of cells with immunosuppressive function (Xiaoyan Li et al. Journal of Immunology 2012). However, whether G-CSF could influence the expression of γδ Tregs remains unknown. The aim of this study was to investigate the effect of G-CSF mobilization on the expression of γδ Tregs. Methods The immunophenotyping of γδ Tregs was analyzed in peripheral blood mononuclear cells (PBMCs) from 20 donors before and after G-CSF mobilization, using flow cytometry. Results Compared with that before mobilization, the proportions of Vδ1 and CD25+ subsets were significantly increased (P=0.012, P=0.032), whereas the Vδ2 proportion was significantly decreased after G-CSF mobilization (P=0.002). The proportions of total γδ T cells, CD27+ and Foxp3+ subsets were similar between the two groups (P=0.133, P=0.110, P=0.780, respectively). In addition, there was a significant increase in the proportions of Foxp3+Vδ1 and CD25+Foxp3+ subsets (P=0.038, P=0.013), and a significant decrease in the proportions of CD27+Vδ2 and CD25+Vδ2 subsets after G-CSF mobilization (P=0.013, P=0.022). The proportions of CD27+γδ T, CD25+γδ T, Foxp3+γδ T, CD25+CD27+, CD27+Foxp3+, CD27+Vδ1, CD25+Vδ1 and Foxp3+Vδ2 subsets were similar before and after G-CSF mobilization (P=0.422, P=0.342, P=0.724, P=0.070, P=0.503, P=0.053, P=0.386 and P=0.097, respectively). We then compared the Foxp3, CD27 and CD25 phenotypes in total γδ T cells, Vδ1 and Vδ2 subsets. We observed a significant increase in the proportion of CD27+Foxp3+ Vδ1 subsets after G-CSF mobilization (P=0.036). The proportion of CD27+Foxp3+γδ T and CD27+Foxp3+Vδ2 subsets before mobilization were similar to that after mobilization (P=0.539, P=0.507). The proportion of CD25+Foxp3+γδ T, CD25+Foxp3+ Vδ1, CD25+Foxp3+Vδ2, CD25+CD27+γδT, CD25+CD27+Vδ1 and CD25+CD27+ Vδ2 subsets were also similar between the two groups (P=0.249, P=0.539, P=0.507, P=0.934, P=0.209 and P=0.061, respectively). Conclusions G-CSF mobilization significantly increased the proportions of Vδ1 subsets, including Foxp3+Vδ1 and CD27+Foxp3+ Vδ1 subsets, whereas decreased the Vδ2 proportion. Disclosures: Li: This work was supported by Grants from National Natural Science Foundation of China (30871091 and 91129720), the Collaborated grant for HK-Macao-TW of Ministry of Science and Technology (2012DFH30060), the Guangdong Science & Technology Project (2012B0506: Research Funding. Liu: It was supported by 863 Program (No. 2011AA020105).: Research Funding; It was supported by National Public Health Grand Research Foundation (Grant No. 201202017), National Natural Science Foundation of China (Grant No.81000231, No.81270647).: Research Funding; It was supported by Science and Technology Program of Guangzhou of China (11A72121174).: Research Funding.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3855-3855
Author(s):  
Li Xuan ◽  
Xiuli Wu ◽  
Min Dai ◽  
Zhenyi Jin ◽  
Xinyu Wang ◽  
...  

Abstract Background Granulocyte colony-stimulating factor (G-CSF) mobilized peripheral blood stem cell has been used more frequently than bone marrow as the source of stem cells in allogeneic hematopoietic stem cell transplantation. Although it contains more mature T cells, neither the incidence nor the severity of acute graft-versus-host disease (aGVHD) is higher compared with bone marrow transplantation. This might be due to the immunoregulatory effects of G-CSF on T cells, including that G-CSF directly modulated via its receptor on T cells or indirectly modulated T cell immune responses via effector cells and cytokines. Recent studies have shown that γδ+ T cells have immunoregulatory function, and might also participate in the pathogenesis of GVHD. However, these mechanisms are not fully understood, and whether G-CSF could influence the immunoregulatory-associated gene expression of γδ+ T cells remains unknown. The aim of this study is to investigate the effect of G-CSF mobilization on the expression of immunoregulatory-associated molecules and G-CSFR gene in the peripheral blood γδ+T cells. Methods Peripheral blood γδ+ T cells were sorted by magnetic activated cell-sorting system. The expression levels of immunoregulatory- associated molecules (Foxp3, CD25, CTLA-4, GITR, TLR8, STAT-1, STAT-3 and RORc) and G-CSFR gene were analyzed in peripheral blood γδ+ T cells from 10 donors before and after G-CSF mobilization, using real-time reverse transcription polymerase chain reaction with SYBR Green I staining. The β2-microglobulin gene was used as an endogenous reference, and the relative mRNA expression level of each gene was evaluated by the 2-ΔCt×100% method. Results The expression levels of Foxp3, CD25, CTLA-4 and GITR genes in peripheral blood γδ+ T cells were similar before and after G-CSF mobilization (P=0.827, P=0.667, P=0.053 and P=0.129). The expression level of TLR8 gene in peripheral blood γδ+ T cells after G-CSF mobilization (0.731% ± 0.350%) was significantly higher than that before mobilization (0.188% ± 0.176%) (P=0.001). The expression level of STAT-1 gene in peripheral blood γδ+ T cells did not differ significantly before and after G-CSF mobilization (P=1.000), while the expression levels of STAT-3, RORc and G-CSFR genes in peripheral blood γδ+ T cells after mobilization (0.536% ± 0.234%, 0.683% ± 0.250%, 8.208% ± 6.175%) were significantly higher than that before mobilization (0.243% ± 0.134%, 0.356% ± 0.179%, 1.947% ± 1.442%) (P=0.003, P=0.003 and P=0.007). Conclusions G-CSF mobilization might increase the expression levels of TLR8, STAT-3, RORc and G-CSFR genes in peripheral blood γδ+T cells. The result suggested that G-CSF might play the role of immunoregulation by affecting γδ+ T cells. Disclosures Xuan: It was supported by National Natural Science Foundation of China (81270647, 81300445, 81200388); National High Technology Research and Development Program of China (863 Program) (2011AA020105): Research Funding. Wu:It was supported by National Natural Science Foundation of China (81270647, 81300445, 81200388); National High Technology Research and Development Program of China (863 Program) (2011AA020105): Research Funding; It was supported by the Technology Plan of Guangdong Province of China (2012B031800403); the project of the Zhujiang Science & Technology Star of Guangzhou city (2013027).: Research Funding. Liu:It was supported by National Natural Science Foundation of China (81270647, 81300445, 81200388); National High Technology Research and Development Program of China (863 Program) (2011AA020105); National Public Health Grand Research Foundation (201202017): Research Funding; It was supported by Natural Science Foundation of Guangdong Province (S2012010009299); the project of health collaborative innovation of Guangzhou city (201400000003-4, 201400000003-1): Research Funding; It was supported by the Technology Plan of Guangdong Province of China (2012B031800403); the project of the Zhujiang Science & Technology Star of Guangzhou city (2013027): Research Funding.


Blood ◽  
1997 ◽  
Vol 89 (5) ◽  
pp. 1629-1634 ◽  
Author(s):  
Marco Mielcarek ◽  
Paul J. Martin ◽  
Beverly Torok-Storb

Abstract The proliferative responsiveness of granulocyte colony-stimulating factor (G-CSF )–mobilized blood was studied in uni-directional mixed leukocyte cultures. Unfractionated mononuclear cells from mobilized blood obtained by leukapheresis at day 4 after initiation of G-CSF (G-PBMC) were hyporesponsive (31.5% ± 9.2% response, P = .003) compared to mononuclear cells obtained from the peripheral blood before administration of G-CSF (preG-PBMC). There was great variability among donors when purified preG- and G-CD4 cells were compared. In eight of 10 donors, G-CD4 cells were equally responsive or moderately hyporesponsive; in two of 10 donors, G-CD4 cells were more strikingly hyporesponsive. CD14 cells derived from leukapheresis products (G-CD14 cells) suppressed alloantigen-induced proliferation by 48.6% ± 7.5% when added to preG-PBMC or preG-CD4 cells at responder-CD14 ratios of 2:1 (P < .001). Suppression was evident (14.4% ± 5.0%) even at responder-CD14 ratios of 8:1 and was largely contact-independent. PreG- and G-CD14 cells had equivalent potency in suppressing proliferative responses. Given that G-CSF–mobilized blood cell grafts contain 50-fold more CD14 cells and only 10-fold more T cells than marrow, we propose that suppression of donor T cells by the large proportion of monocytes present in leukapheresis products could contribute to the unexpectedly low incidence and severity of graft-versus-host disease after peripheral blood stem cell transplantation.


Blood ◽  
1997 ◽  
Vol 89 (5) ◽  
pp. 1629-1634 ◽  
Author(s):  
Marco Mielcarek ◽  
Paul J. Martin ◽  
Beverly Torok-Storb

The proliferative responsiveness of granulocyte colony-stimulating factor (G-CSF )–mobilized blood was studied in uni-directional mixed leukocyte cultures. Unfractionated mononuclear cells from mobilized blood obtained by leukapheresis at day 4 after initiation of G-CSF (G-PBMC) were hyporesponsive (31.5% ± 9.2% response, P = .003) compared to mononuclear cells obtained from the peripheral blood before administration of G-CSF (preG-PBMC). There was great variability among donors when purified preG- and G-CD4 cells were compared. In eight of 10 donors, G-CD4 cells were equally responsive or moderately hyporesponsive; in two of 10 donors, G-CD4 cells were more strikingly hyporesponsive. CD14 cells derived from leukapheresis products (G-CD14 cells) suppressed alloantigen-induced proliferation by 48.6% ± 7.5% when added to preG-PBMC or preG-CD4 cells at responder-CD14 ratios of 2:1 (P < .001). Suppression was evident (14.4% ± 5.0%) even at responder-CD14 ratios of 8:1 and was largely contact-independent. PreG- and G-CD14 cells had equivalent potency in suppressing proliferative responses. Given that G-CSF–mobilized blood cell grafts contain 50-fold more CD14 cells and only 10-fold more T cells than marrow, we propose that suppression of donor T cells by the large proportion of monocytes present in leukapheresis products could contribute to the unexpectedly low incidence and severity of graft-versus-host disease after peripheral blood stem cell transplantation.


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 ◽  
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.


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 ◽  
1997 ◽  
Vol 90 (1) ◽  
pp. 453-463 ◽  
Author(s):  
Defu Zeng ◽  
Sussan Dejbakhsh-Jones ◽  
Samuel Strober

The feasibility of transplanting peripheral blood mononuclear cells (PBMC) from granulocyte colony-stimulating factor (G-CSF)–treated normal human donors to myeloablated allogeneic hosts has been demonstrated recently. The current work examined the ability of recombinant G-CSF to alter peripheral blood T-cell function and graft-versus-host disease (GVHD) in a murine model of allogeneic G-CSF–mobilized PBMC transplantation. Administration of recombinant G-CSF to C57BL/Ka mice markedly increased the capacity of PBMC to reconstitute lethally irradiated syngeneic hosts. T- and B-lineage lymphocytes were depleted about 10-fold in the bone marrow of the treated mice, and the T-cell yield in the blood was increased about fourfold. The ability of PBMC or purified CD4+ and CD8+ T cells to induce acute lethal GVHD in irradiated BALB/c mice was reduced after the administration of G-CSF. This was associated with decreased secretion of interferonγ and interleukin-2 (IL-2) and an increased secretion of IL-4. The donor cell inoculum, which was most successful in the rescue of irradiated allogeneic hosts, was the low-density fraction of PBMC from G-CSF–treated mice. These low-density cells were enriched for CD4−CD8−NK1.1+ T cells and secreted about 10-fold more IL-4 than the unfractionated cells from the G-CSF–treated donors.


Sign in / Sign up

Export Citation Format

Share Document