Granulocyte colony-stimulating factor induced T-cell hyporesponsiveness via modulation of CD177+S100Ahi neutrophils in unexplained recurrent pregnancy loss

Ping-Fen Li , Xue Zhang , Peng-Sheng Zheng

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70508

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70508 DOI: 10.1002/ctm2.70508
RESEARCH ARTICLE

Granulocyte colony-stimulating factor induced T-cell hyporesponsiveness via modulation of CD177+S100Ahi neutrophils in unexplained recurrent pregnancy loss

Author information +
History +
PDF

Abstract

Background: Numerous studies have demonstrated the promising efficacy of granulocyte colony-stimulating factor (G-CSF) in the treatment of couples with unexplained recurrent pregnancy loss (URPL) during early pregnancy. While neutrophils are recognised as the main effectors mediating immunoregulation, their G-CSF-mobilised phenotype and mechanisms regulating maternal–fetal immunity remain unclear.

Methods: Single-cell RNA sequencing (scRNA-seq) and single-cell T-cell receptor sequencing (scTCR-seq) were conducted to uncover the immune reconstitution dynamics of peripheral blood under G-CSF stimulation. Integrative analysis of transcriptomic-proteomic profiles with functional validation revealed a unique immunomodulatory neutrophil population. Further, we used spatial transcriptomics, flow cytometry and immunohistochemistry to explore the spatial distribution characteristics of this population at the maternal–fetal interface, and validated its therapeutic efficacy in animal models.

Results: G-CSF-mobilised peripheral blood (G-PB) displayed immune hyporesponsiveness. Unique neutrophils expressing high levels of CD177 and the S100A gene family expanded substantially in response to G-CSF. These neutrophils exhibited a comparatively immature morphology and impaired T-cell responses via contact-dependent arginase 1 release, as well as upregulation of T-cell immune checkpoints. A reduction of CD177+S100Ahi neutrophils was observed in both peripheral blood and decidua of URPL patients relative to healthy pregnant women. Functional validation in abortion-prone murine models confirmed that exogenous supplementation of G-CSF or adoptive transfer of CD177+S100Ahi neutrophils could successfully improve the pregnancy outcomes.

Conclusion: G-CSF played a crucial regulatory role in improving pregnancy outcomes by selectively expanding CD177+S100Ahi neutrophils with polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) properties, providing a solid theoretical foundation for the treatment of patients with URPL using G-CSF.

Keywords

G-CSF / PMN-MDSCs / scRNA-seq / scTCR-seq / URPL

Cite this article

Download citation ▾
Ping-Fen Li, Xue Zhang, Peng-Sheng Zheng. Granulocyte colony-stimulating factor induced T-cell hyporesponsiveness via modulation of CD177+S100Ahi neutrophils in unexplained recurrent pregnancy loss. Clinical and Translational Medicine, 2025, 15(10): e70508 DOI:10.1002/ctm2.70508

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Mor G. Introduction to the immunology of pregnancy. Immunol Rev. 2022; 308: 5-8.

[2]

Xu L, Li Y, Sang Y, Li DJ, Du M. Crosstalk between trophoblasts and decidual immune cells: the cornerstone of maternal-fetal immunotolerance. Front Immunol. 2021; 12: 642392.

[3]

Bender Atik R, Christiansen OB, Elson J, et al. ESHRE guideline: recurrent pregnancy loss. Human Reprod Open. 2018; 2018: hoy004.

[4]

Definitions of infertility and recurrent pregnancy loss: a committee opinion. Fertil Steril. 2020; 113: 533-535.

[5]

Regan L, Rai R. Epidemiology and the medical causes of miscarriage. Best Pract Res Clin Obstet Gynaecol. 2000; 14: 839-854.

[6]

Cramer DW, Wise LA. The epidemiology of recurrent pregnancy loss. Semin Reprod Med. 2000; 18: 331-339.

[7]

Eftekhar M, Naghshineh E, Khani P. Role of granulocyte colony-stimulating factor in human reproduction. J Res Med Sci. 2018; 23: 7.

[8]

Cavalcante MB, Costa Fda S, Barini R, Araujo Júnior E. Granulocyte colony-stimulating factor and reproductive medicine: a review. Iran J Reprod Med. 2015; 13: 195-202.

[9]

Ding J, Wang J, Cai X, et al. Granulocyte colony-stimulating factor in reproductive-related disease: function, regulation and therapeutic effect. Biomed Pharmacother. 2022; 150: 112903.

[10]

Gao P, Zha Y, Wei L, et al. A vehicle for communication between trophoblasts and macrophages which may cause problems in recurrent spontaneous abortion. Placenta. 2022; 121: 164-172.

[11]

Scarpellini F, Klinger FG, Rossi G, Sbracia M. Immunohistochemical study on the expression of G-CSF, G-CSFR, VEGF, VEGFR-1, Foxp3 in first trimester trophoblast of recurrent pregnancy loss in pregnancies treated with G-CSF and controls. Int J Mol Sci. 2019; 21: 285.

[12]

Scarpellini F, Sbracia M. Use of granulocyte colony-stimulating factor for the treatment of unexplained recurrent miscarriage: a randomised controlled trial. Hum Reprod. 2009; 24: 2703-2708.

[13]

Santjohanser C, Knieper C, Franz C, et al. Granulocyte-colony stimulating factor as treatment option in patients with recurrent miscarriage. Arch Immunol Ther Exp (Warsz). 2013; 61: 159-164.

[14]

Mu F, Huang J, Zeng X, Liu L, Wang F. Efficacy and safety of recombinant human granulocyte colony-stimulating factor in patients with unexplained recurrent spontaneous abortion: a systematic review and meta-analysis. J Reprod Immunol. 2023; 156: 103830.

[15]

Shao Q, Liu X, Huang Y, Chen X, Wang H. Human decidual stromal cells in early pregnancy induce functional re-programming of monocyte-derived dendritic cells via crosstalk between G-CSF and IL-1β. Front Immunol. 2020; 11: 574270.

[16]

Li W, Zhang X, Chen Y, et al. G-CSF is a key modulator of MDSC and could be a potential therapeutic target in colitis-associated colorectal cancers. Protein Cell. 2016; 7: 130-140.

[17]

Abrams SI, Waight JD. Identification of a G-CSF-granulocytic MDSC axis that promotes tumour progression. Oncoimmunology. 2012; 1: 550-551.

[18]

Schmid KT, Höllbacher B, Cruceanu C, et al. scPower accelerates and optimizes the design of multi-sample single cell transcriptomic studies. Nat Commun. 2021; 12: 6625.

[19]

Hu XE, Yang P, Chen S, et al. Clinical and biological heterogeneities in triple-negative breast cancer reveals a non-negligible role of HER2-low. Breast Cancer Res. 2023; 25: 34.

[20]

Li QS, Zheng PS. ESRRB inhibits the TGFβ signaling pathway to drive cell proliferation in cervical cancer. Cancer Res. 2023; 83: 3095-3114.

[21]

Lei D, Yang WT, Zheng PS. HOXB4 inhibits the proliferation and tumourigenesis of cervical cancer cells by downregulating the activity of Wnt/β-catenin signaling pathway. Cell Death Dis. 2021; 12: 105.

[22]

Wang G, Wu S, Xiong Z, Qu H, Fang X, Bao Y. CROST: a comprehensive repository of spatial transcriptomics. Nucleic Acids Res. 2024; 52: D882-D890.

[23]

Wu Y, Su K, Zhang Y, et al. A spatiotemporal transcriptomic atlas of mouse placentation. Cell Discov. 2024; 10: 110.

[24]

Yang M, Ong J, Meng F, et al. Spatiotemporal insight into early pregnancy governed by immune-featured stromal cells. Cell. 2023; 186: 4271-4288.e24.

[25]

Wang J, Yang J, Yan Y, et al. Effect of adoptive transfer of CD4(+)CD25(+)Foxp3(+) Treg induced by trichostatin A on the prevention of spontaneous abortion. J Reprod Immunol. 2019; 131: 30-35.

[26]

Jia W, Ma L, Yu X, et al. Human CD56(+)CD39(+) dNK cells support fetal survival through controlling trophoblastic cell fate: immune mechanisms of recurrent early pregnancy loss. Natl Sci Rev. 2024; 11: nwae142.

[27]

Maneta E, Fultang L, Taylor J, et al. G-CSF induces CD15(+) CD14(+) cells from granulocytes early in the physiological environment of pregnancy and the cancer immunosuppressive microenvironment. Clin Transl Immunol. 2022; 11: e1395.

[28]

Xie X, Shi Q, Wu P, et al. Single-cell transcriptome profiling reveals neutrophil heterogeneity in homeostasis and infection. Nat Immunol. 2020; 21: 1119-1133.

[29]

Silvestre-Roig C, Fridlender ZG, Glogauer M, Scapini P. Neutrophil diversity in health and disease. Trends Immunol. 2019; 40: 565-583.

[30]

Schneider WM, Chevillotte MD, Rice CM. Interferon-stimulated genes: a complex web of host defenses. Annu Rev Immunol. 2014; 32: 513-545.

[31]

Ding J, Maxwell A, Adzibolosu N, et al. Mechanisms of immune regulation by the placenta: role of type I interferon and interferon-stimulated genes signaling during pregnancy. Immunol Rev. 2022; 308: 9-24.

[32]

Evrard M, Kwok IWH, Chong SZ, et al. Developmental analysis of bone marrow neutrophils reveals populations specialized in expansion, trafficking, and effector functions. Immunity. 2018; 48: 364-379.e8.

[33]

Ikeda N, Kubota H, Suzuki R, et al. The early neutrophil-committed progenitors aberrantly differentiate into immunoregulatory monocytes during emergency myelopoiesis. Cell Rep. 2023; 42: 112165.

[34]

Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021; 21: 485-498.

[35]

von Wulffen M, Luehrmann V, Robeck S, et al. S100A8/A9-alarmin promotes local myeloid-derived suppressor cell activation restricting severe autoimmune arthritis. Cell Rep. 2023; 42: 113006.

[36]

Li K, Shi H, Zhang B, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021; 6: 362.

[37]

Zhou J, Nefedova Y, Lei A. Neutrophils and PMN-MDSC: their biological role and interaction with stromal cells. Semin Immunol. 2018; 35: 19-28.

[38]

Nguyen CT, Furuya H, Das D, et al. Peripheral γδ T cells regulate neutrophil expansion and recruitment in experimental psoriatic arthritis. Arthritis Rheumatol (Hoboken, NJ). 2022; 74: 1524-1534.

[39]

Pelletier M, Maggi L, Micheletti A, et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood. 2010; 115: 335-343.

[40]

Hirschhorn D, Budhu S, Kraehenbuehl L, et al. T-cell immunotherapies engage neutrophils to eliminate tumour antigen escape variants. Cell. 2023; 186: 1432-1447.e17.

[41]

Aoki H, Shichino S, Matsushima K, Ueha S. Revealing clonal responses of tumour-reactive T-cells through T cell receptor repertoire analysis. Front Immunol. 2022; 13: 807696.

[42]

Pai JA, Satpathy AT. High-throughput and single-cell T-cell receptor sequencing technologies. Nat Methods. 2021; 18: 881-892.

[43]

Tinoco R, Otero DC, Takahashi AA, Bradley LM. PSGL-1: a new player in the immune checkpoint landscape. Trends Immunol. 2017; 38: 323-335.

[44]

Tinoco R, Carrette F, Barraza ML, et al. PSGL-1 is an immune checkpoint regulator that promotes T cell exhaustion. Immunity. 2016; 44: 1190-1203.

[45]

Collins AS, Ahmed S, Napoletano S, et al. Hepatitis C virus (HCV)-induced suppressor of cytokine signaling (SOCS) 3 regulates proinflammatory TNF-α responses. J Leukocyte Biol. 2014; 96: 255-263.

[46]

Jia L, Shi Y, Wen Y, Li W, Feng J, Chen C. The roles of TNFAIP2 in cancers and infectious diseases. J Cell Mol Med. 2018; 22: 5188-5195.

[47]

Huyghe J, Priem D, Bertrand MJM. Cell death checkpoints in the TNF pathway. Trends Immunol. 2023; 44: 628-643.

[48]

Mehta HM, Corey SJ. G-CSF, the guardian of granulopoiesis. Semin Immunol. 2021; 54: 101515.

[49]

Martin KR, Wong HL, Witko-Sarsat V, Wicks IP. G-CSF—a double edge sword in neutrophil mediated immunity. Semin Immunol. 2021; 54: 101516.

[50]

Bennett JA, Rao VS, Mitchell MS. Systemic bacillus Calmette-Guérin (BCG) activates natural suppressor cells. Proc Natl Acad Sci U.S.A. 1978; 75: 5142-5144.

[51]

Zheng ZM, Yang HL, Lai ZZ, et al. Myeloid-derived suppressor cells in obstetrical and gynecological diseases. Am J Reprod Immunol. 2020; 84: e13266.

[52]

Köstlin N, Kugel H, Spring B, et al. Granulocytic myeloid derived suppressor cells expand in human pregnancy and modulate T-cell responses. Eur J Immunol. 2014; 44: 2582-2591.

[53]

Zhao AM, Xu HJ, Kang XM, Zhao AM, Lu LM. New insights into myeloid-derived suppressor cells and their roles in feto-maternal immune cross-talk. J Reprod Immunol. 2016; 113: 35-41.

[54]

Hegde S, Leader AM, Merad M. MDSC: markers, development, states, and unaddressed complexity. Immunity. 2021; 54: 875-884.

[55]

Marini O, Costa S, Bevilacqua D, et al. Mature CD10(+) and immature CD10(−) neutrophils present in G-CSF-treated donors display opposite effects on T cells. Blood. 2017; 129: 1343-1356.

[56]

Li C, Chen C, Kang X, et al. Decidua-derived granulocyte macrophage colony-stimulating factor induces polymorphonuclear myeloid-derived suppressor cells from circulating CD15+ neutrophils. Hum Reprod. 2020; 35: 2677-2691.

[57]

Huo Y, Wu L, Pang A, et al. Single-cell dissection of human hematopoietic reconstitution after allogeneic hematopoietic stem cell transplantation. Sci Immunol. 2023; 8: eabn6429.

[58]

Zhao X, Peng T, Cao X, et al. In vivo G-CSF treatment activates the GR-SOCS1 axis to suppress IFN-γ secretion by natural killer cells. Cell Rep. 2022; 40: 111342.

[59]

Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol. 2018; 19: 108-119.

[60]

Shi M, Chen Z, Chen M, et al. Continuous activation of polymorphonuclear myeloid-derived suppressor cells during pregnancy is critical for fetal development. Cell Mol Immunol. 2021; 18: 1692-1707.

[61]

Lauret Marie, Joseph E, Laheurte C, et al. Immunoregulation and clinical implications of ANGPT2/TIE2(+) M-MDSC signature in non-small cell lung cancer. Cancer Immunol Res. 2020; 8: 268-279.

[62]

Perez C, Botta C, Zabaleta A, et al. Immunogenomic identification and characterization of granulocytic myeloid-derived suppressor cells in multiple myeloma. Blood. 2020; 136: 199-209.

[63]

Bernard V, Semaan A, Huang J, et al. Single-cell transcriptomics of pancreatic cancer precursors demonstrates epithelial and microenvironmental heterogeneity as an early event in neoplastic progression. 2019; 25: 2194-2205.

[64]

Condamine T, Dominguez GA, Youn JI, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 2016; 1: aaf8943.

[65]

Huang C, Fan X, Shen Y, Shen M, Yang L. Neutrophil subsets in noncancer liver diseases: cellular crosstalk and therapeutic targets. Eur J Immunol. 2023; 53: e2250324.

[66]

Minns D, Smith KJ, Hardisty G, Rossi AG, Gwyer Findlay E. The outcome of neutrophil-T cell contact differs depending on activation status of both cell types. Front Immunol. 2021; 12: 633486.

[67]

Michaeli J, Shaul ME, Mishalian I, et al. Tumour-associated neutrophils induce apoptosis of non-activated CD8 T-cells in a TNFα and NO-dependent mechanism, promoting a tumour-supportive environment. Oncoimmunology. 2017; 6: e1356965.

[68]

Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017; 5: 3-8.

[69]

Bert S, Nadkarni S, Perretti M. Neutrophil-T-cell crosstalk and the control of the host inflammatory response. Immunol Rev. 2023; 314: 36-49.

[70]

Geiger R, Rieckmann JC, Wolf T, et al. L-Arginine modulates T cell metabolism and enhances survival and anti-tumour activity. Cell. 2016; 167: 829-842.e13.

[71]

Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol. 2022; 19: 1079-1101.

[72]

Munder M, Schneider H, Luckner C, et al. Suppression of T-cell functions by human granulocyte arginase. Blood. 2006; 108: 1627-1634.

[73]

Rodriguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC. Regulation of T-cell receptor CD3zeta chain expression by L-arginine. J Biol Chem. 2002; 277: 21123-21129.

[74]

Feldmeyer N, Wabnitz G, Leicht S, et al. Arginine deficiency leads to impaired cofilin dephosphorylation in activated human T lymphocytes. Int Immunol. 2012; 24: 303-313.

[75]

Cemerski S, Cantagrel A, Van Meerwijk JP, Romagnoli P. Reactive oxygen species differentially affect T-cell receptor-signaling pathways. J Biol Chem. 2002; 277: 19585-19593.

[76]

Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020; 21: 363-383.

[77]

Franchina DG, Dostert C, Brenner D. Reactive oxygen species: involvement in T cell signaling and metabolism. Trends Immunol. 2018; 39: 489-502.

[78]

Chen D, Wang W, Wu L, et al. Single-cell atlas of peripheral blood mononuclear cells from pregnant women. Clin Transl Med. 2022; 12: e821.

[79]

Li C, Zhang X, Kang X, et al. Upregulated TRAIL and reduced DcR2 mediate apoptosis of decidual PMN-MDSC in unexplained recurrent pregnancy loss. Front Immunol. 2020; 11: 1345.

[80]

Qin D, Xu H, Chen Z, et al. The peripheral and decidual immune cell profiles in women with recurrent pregnancy loss. Front Immunol. 2022; 13: 994240.

[81]

Sojka DK, Yang L, Yokoyama WM. Uterine natural killer cells. Front Immunol. 2019; 10: 960.

[82]

Tong X, Gao M, Du X, et al. Analysis of uterine CD49a(+) NK cell subsets in menstrual blood reflects endometrial status and association with recurrent spontaneous abortion. Cell Mol Immunol. 2021; 18: 1838-1840.

[83]

van der Molen RG, Schutten JH, van Cranenbroek B, et al. Menstrual blood closely resembles the uterine immune micro-environment and is clearly distinct from peripheral blood. Hum Reprod. 2014; 29: 303-314.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

9

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/