Myeloid-Derived Suppressor Cells (MDSC) Are Effectors of Bone Marrow Suppression in Lower Risk Myelodysplastic Syndromes (MDS).

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 597-597 ◽  
Author(s):  
Sheng Wei ◽  
Xianghong Chen ◽  
Kathy Rocha ◽  
Dahui Qi ◽  
Jianguo Tao ◽  
...  

Abstract Abstract 597 The acquisition of genetic abnormalities that lead to ineffective hematopoiesis is a characteristic of MDS. This event is mediated in part by an interaction of inflammatory intermediates with the bone marrow (BM) microenvironment; however, the mechanism by which chromosomal damage occurs to the stem/progenitor compartment (HSC/HPC) is unknown. We have identified a unique population of myeloid derived suppressor cells (MDSC) bearing a LIN-HLA-DR-CD33+ surface phenotype. These MDSC elicit hematopoietic suppression through the elaboration of nitric oxide, arginase, and inflammatory cytokines. This study found that MDSCs accumulated in excess in the BM of patients with lower risk MDS (mean, 32.32% ± 13.3; n=12) compared to BM from healthy age matched controls (mean 2.1%±0.9; n=8) and non-MDS cancer patients (n=8 mean 2.06%±1.5 p<0.0001). Fluorescence in situ hybridization (FISH) was performed to determine whether MDSCs represent a distinct cell population from the abnormal MDS clone. MDSCs from BM of 5 patients having chromosomal abnormality were separated by FACS sorting based on LIN-HLA-DR-CD33+ phenotype and the presence of chromosomal abnormalities was determined in this population and compared to non-MDSCs. Monosomy 7 and deletion of 5q chromosomal abnormalities resided within the non-MDSC hematopoietic compartment. This indicates that MDSCs in low risk MDS patients may represent a unique cell population from the HPCs with clonal potential. Furthermore, the key cytokines involved in MDSC suppressive function, TGFβ, VEGF and IL-10, were higher in MDSCs isolated from MDS patients compared to controls. Using four-color immunostaining, we discovered that MDSCs are capable of direct cytotoxicity against autologous erythroid precursors (CD71+ and CD235a+), as evidenced by increased polarized granule mobilization toward the site of cellular contact. Moreover, it was observed that the accumulation of MDSC in the BM from MDS patients has an impact on hematopoietic differentiation. In the presence of MDSCs, the formation of BFU-E (burst forming unit erythroid) was significantly suppressed in all patients tested (n=6). In contrast depletion of the MDSC by FACS sorting reconstituted the formation of BFU-E (21± 4.2 in MDSC depleted bone marrow verse 0.8± 0.6 in unsorted BM cells, respectively). These data strongly suggest that the presence of MDSCs in the BM microenvironment of MDS patients contribute to suppression of HPC development. Based on these findings, a novel form of adaptive immunotherapy based on the induction of MDSC maturation can be envisioned. DAP12 (DNAX-activating protein of 12kDa) is an adaptor protein that mediates signaling of dentritic cell and monocyte maturation. In order to determine if DAP12 signaling induces MDSC maturation and reversal of suppressive function, a genetically modified, constitutively activated form was introduced into BM mononuclear cells (BM-MNC) using both recombinant adenoviral and lentiviral gene transduction. Infection of BM-MNC from MDS patients with constitutively active DAP12 increased expression of maturation surface markers CD14, CD15 and HLA-DR and increased BFU-E colony formation (31± 2.1 verse control 7±0.7) after 14 days. These results suggest that active signaling through DAP12 has potential therapeutic implications in MDS by driving maturation of immature myeloid cells reversing the inflammatory changes that contribute to malignant transformation. In conclusion, our previously unknown findings have begun to elucidate whether the presence of MDSCs in the BM microenvironment of MDS patients contributes to the pathogenesis of MDS by providing a suppressive microenvironment and repress BM maturation associated with impaired HSC/HPC development. Additionally, DAP12 may be a potential therapeutic pathway in which to stimulate MDSC maturation, reversing the suppressive effects on HSC/HPC development. Disclosures: No relevant conflicts of interest to declare.

2016 ◽  
Vol 130 (4) ◽  
pp. 259-271 ◽  
Author(s):  
Veronica I. Landoni ◽  
Daiana Martire-Greco ◽  
Nahuel Rodriguez-Rodrigues ◽  
Paula Chiarella ◽  
Pablo Schierloh ◽  
...  

LPS-induced immunosuppression, mimicking the state observed in patients with late sepsis, induced in bone marrow a population of myeloid-derived suppressor cells (Gr-1+ CD11b+) with the ability to inhibit T-cell responses and migrate to lymph nodes to exert their suppressive function.


2015 ◽  
Vol 35 (1) ◽  
pp. 292-304 ◽  
Author(s):  
Yan-ge Wang ◽  
Xin Xiong ◽  
Zhu-yue Chen ◽  
Kan-ling Liu ◽  
Jin-hua Yang ◽  
...  

Aim: The aim of this study was to explore whether the circulating frequency and function of myeloid-derived suppressor cells (MDSCs) are altered in patients with acute coronary syndrome (ACS). Methods: The frequency of MDSCs in peripheral blood was determined by flow cytometry, and mRNA expression in purified MDSCs was analyzed by real-time reverse transcription polymerase chain reaction (RT-PCR). The suppressive function of MDSCs isolated from different groups was also determined. The plasma levels of certain cytokines were determined using Bio-Plex Pro™ Human Cytokine Assays. Results: The frequency of circulating CD14+HLA-DR-/low MDSCs; arginase-1 (Arg-1) expression; and plasma levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and IL-33 were markedly increased in ACS patients compared to stable angina (SA) or control patients. Furthermore, MDSCs from ACS patients were more potent suppressors of T-cell proliferation and IFN-γ production than those from the SA or control groups at ratios of 1:4 and 1:2; this effect was partially mediated by Arg-1. In addition, the frequency of MDSCs was positively correlated with plasma levels of IL-6, IL-33, and TNF-α. Conclusions: We observed an increased frequency and suppressive function of MDSCs in ACS patients, a result that may provide insights into the mechanisms involved in ACS.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A536-A536
Author(s):  
Juan Dong ◽  
Cassandra Gilmore ◽  
Hieu Ta ◽  
Keman Zhang ◽  
Sarah Stone ◽  
...  

BackgroundV-domain immunoglobulin suppressor of T cell activation (VISTA) is a B7 family inhibitory immune checkpoint protein and is highly expressed on myeloid cells and T cells.1 VISTA acts as both an inhibitory ligand when expressed on antigen-presenting cells and a receptor when expressed on T cells. Our recent study has shown that VISTA is a myeloid cell-specific immune checkpoint and that blocking VISTA can reprogram suppressive myeloid cells and promote a T cell-stimulatory tumor microenvironment.2 In this study, we further demonstrate that VISTA blockade directly alters the differentiation and the suppressive function of myeloid-derived suppressor cells (MDSC).MethodsFlow cytometry was performed to examine VISTA expression on MDSCs in multiple murine tumor models including the B16BL6 melanoma model, MC38 colon cancer model, and the KPC pancreatic cancer models. To examine the role of VISTA in controlling the differentiation and suppressive function of MDSCs, we cultured wild type (WT) and VISTA.KO bone marrow progenitor cells with GM-CSF and IL-6 to induce BM -derived MDSCs.ResultsOur preliminary results show that VISTA is highly expressed on M-MDSCs in B16BL6, MC38 and KPC tumors. In BM-derived MDSCs, VISTA deletion significantly altered the signaling pathways and the differentiation of MDSCs. Multiple inflammatory signaling pathways were downregulated in VISTA KO MDSCs, resulting in decreased production of cytokines such as IL1 and chemokines such as CCL2/4/9, as well as significantly impaired their ability to suppress the activation of CD8+ T cells. The loss of suppressive function in VISTA KO MDSCs is correlated with significantly reduced expression of iNOS. To validate the results from BM-MDSCs, we sorted CD11b+CD11c-Ly6C+Ly6G- M-MDSCs and CD11b+CD11c-Ly6G+ G-MDSCs from B16BL6 tumor tissues and tested the ability of a VISTA-blocking mAb to reverse the suppressive effects of tumor-derived MDSCs. Our results show that blocking VISTA impaired the suppressive function of tumor-derived M-MDSC but not G-MDSCs.ConclusionsTaken together, these results demonstrate a crucial role of VISTA in regulating the differentiation and function of MDSCs, and that blocking VISTA abolishes MDSC-mediated T cell suppression, thereby boosting.Ethics ApprovalAll in vivo studies were reviewed and approved by Institutional Animal Care and Use Committee (Approval number 2019-2142).ReferencesXu W, Hire T, Malarkannan, S. et al. The structure, expression, and multifaceted role of immune-checkpoint protein VISTA as a critical regulator of anti-tumor immunity, autoimmunity, and inflammation. Cell Mol Immunol 2018;15:438–446.Xu W, Dong J, Zheng Y, et al. Immune-checkpoint protein VISTA regulates antitumor immunity by controlling myeloid cell-mediated inflammation and immunosuppression. Cancer Immunol Res 2019;7:1497–510.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Clémence Marais ◽  
Caroline Claude ◽  
Nada Semaan ◽  
Ramy Charbel ◽  
Simon Barreault ◽  
...  

Abstract Background De-regulated host response to severe coronavirus disease 2019 (COVID-19), directly referring to the concept of sepsis-associated immunological dysregulation, seems to be a strong signature of severe COVID-19. Myeloid cells phenotyping is well recognized to diagnose critical illness-induced immunodepression in sepsis and has not been well characterized in COVID-19. The aim of this study is to review phenotypic characteristics of myeloid cells and evaluate their relations with the occurrence of secondary infection and mortality in patients with COVID-19 admitted in an intensive care unit. Methods Retrospective analysis of the circulating myeloid cells phenotypes of adult COVID-19 critically ill patients. Phenotyping circulating immune cells was performed by flow cytometry daily for routine analysis and twice weekly for lymphocytes and monocytes subpopulations analysis, as well as monocyte human leukocyte antigen (mHLA)-DR expression. Results Out of the 29 critically ill adult patients with severe COVID-19 analyzed, 12 (41.4%) developed secondary infection and six patients died during their stay. Monocyte HLA-DR kinetics was significantly different between patients developing secondary infection and those without, respectively, at day 5–7 and 8–10 following admission. The monocytes myeloid-derived suppressor cells to total monocytes ratio was associated with 28- and 60-day mortality. Those myeloid characteristics suggest three phenotypes: hyperactivated monocyte/macrophage is significantly associated with mortality, whereas persistent immunodepression is associated with secondary infection occurrence compared to transient immunodepression. Conclusions Myeloid phenotypes of critically ill COVID-19 patients may be associated with development of secondary infection, 28- and 60-day mortality.


Author(s):  
Caio César Barbosa Bomfim ◽  
Eduardo Pinheiro Amaral ◽  
Igor Santiago-Carvalho ◽  
Gislane Almeida Santos ◽  
Érika Machado Salles ◽  
...  

Abstract Background The role of myeloid-derived suppressor cells (MDSCs) in severe tuberculosis patients who suffer from uncontrolled pulmonary inflammation caused by hypervirulent mycobacterial infection remains unclear. Methods This issue was addressed using C57BL/6 mice infected with highly virulent Mycobacterium bovis strain MP287/03. Results CD11b +GR1 int population increased in the bone marrow, blood and lungs during advanced disease. Pulmonary CD11b +GR1 int (Ly6G intLy6C int) cells showed granularity similar to neutrophils and expressed immature myeloid cell markers. These immature neutrophils harbored intracellular bacilli and were preferentially located in the alveoli. T cell suppression occurred concomitantly with CD11b +GR1 int cell accumulation in the lungs. Furthermore, lung and bone-marrow GR1 + cells suppressed both T cell proliferation and IFN-γ production in vitro. Anti-GR1 therapy given when MDSCs infiltrated the lungs prevented expansion and fusion of primary pulmonary lesions and the development of intragranulomatous caseous necrosis, along with increased mouse survival and partial recovery of T cell function. Lung bacterial load was reduced by anti-GR1 treatment, but mycobacteria released from the depleted cells proliferated extracellularly in the alveoli, forming cords and clumps. Conclusions Granulocytic MDSCs massively infiltrate the lungs during infection with hypervirulent mycobacteria, promoting bacterial growth and the development of inflammatory and necrotic lesions, and are promising targets for host-directed therapies.


Sign in / Sign up

Export Citation Format

Share Document