T Helper (Th) Cell Profiles in Pregnancy and Recurrent Pregnancy Losses: Th1/Th2/Th9/Th17/Th22/Tfh Cells

Wenjuan Wang, Nayoung Sung, Alice Gilman-Sachs, Joanne Kwak-Kim, Wenjuan Wang, Nayoung Sung, Alice Gilman-Sachs, Joanne Kwak-Kim

Abstract

During pregnancy, various immune effectors and molecules participating in the immune-microenvironment establish specific maternal tolerance toward the semi-allogeneic fetus. Activated maternal immune effectors by the trophoblast antigens, such as T helper (Th), T cytotoxic (Tc), T regulatory (Treg), and B cells, are involved in the regulation of adaptive immunity. Recognition of active signal through the T cell receptors stimulate the differentiation of naive CD3+CD4+ T cells into specific T cell subsets, such as Th1, Th2, Th9, Th17, Th22, and follicular Th cells (Tfh). Each of these subsets has a significant and distinct role in human pregnancy. Th1 immunity, characterized by immune-inflammatory responses, becomes dominant during the peri-implantation period, and the "controlled" Th1 immunity benefits the invading trophoblasts rather than harm. Quickly after the placental implantation, the early inflammatory Th1 immunity is shifted to the Th2 anti-inflammatory immune responses. The predominant Th2 immunity, which overrules the Th1 immunity at the placental implantation site, protects a fetus by balancing Th1 immunity and accommodate fetal and placental development. Moreover, Treg and Th9 cells regulate local inflammatory immune responses, potentially detrimental to the fetus. Th17 cells induce protective immunity against extracellular microbes during pregnancy. However, excessive Th17 immunity may induce uncontrolled neutrophil infiltration at the maternal-fetal interface. Other Th cell subsets such as Tfh cells, also contribute to pregnancy by setting up favorable humoral immunity during pregnancy. However, dysregulation of Th cell immunity during pregnancy may result in obstetrical complications, such as recurrent pregnancy losses (RPL) and preeclampsia (PE). With this review, we intend to deliver a comprehensive overview of CD4+ Th cell subsets, including Th1, Th2, Th9, Th17, Th22, and Tfh cells, in human pregnancy by reviewing their roles in normal and pathological pregnancies.

Keywords: Th1 cell; Th17 cells; Th2 cells; Th22 cells; Th9 cells; pregnancy; recurrent pregnancy loss.

Copyright © 2020 Wang, Sung, Gilman-Sachs and Kwak-Kim.

Figures

FIGURE 1
FIGURE 1
Classical and alternative Th17 and Th22 subsets are differentiated from the naïve CD4+ T cells based on differentiation cytokines. Activated Th17 and Th22 cells have critical roles for the maintenance of pregnancy while establishing inflammation and immunity against extracellular pathogens at the maternal-fetal junction.
FIGURE 2
FIGURE 2
Timely shifting of Th1/Th2 and Treg/Th17 balance is of utmost importance for a successful pregnancy. During the implantation phase, Th1 cells are infiltrated at the decidua and regulate trophoblast invasion by participating in tissue remodeling and angiogenesis. Th17 and Th22 cells are present in the decidua but not enriched. Th17 cells involve in neutrophil infiltration and induction of protective immunity against extracellular microbes in the uterus. Th22 cells also protect trophoblast cells from pathogens and inflammatory immune responses from intrauterine infection. Decreased Th22 cells induce dysfunction of decidual stromal cells and NK cells. Hence, Th22 cells are keenly associated with Th1/Th2 balance. Treg and Th2 cells repress excessive Th1 and Th17 immunity. When the implantation phase is over, Th2 and Th9 cells become dominant and promote allograft tolerance. Tfh cells are usually enriched at late gestation, favoring humoral immunity and, thus, balancing Th1/Th2 immune responses.

References

    1. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol. (1986) 136:2348–57.
    1. Wegmann TG, Lin H, Guilbert L, Mosmann TR. Bidirectional cytokine interactions in the maternal-fetal relationship: is successful pregnancy a TH2 phenomenon? Immunol Today. (1993) 14:353–6. 10.1016/0167-5699(93)90235-D
    1. Piccinni MP, Beloni L, Livi C, Maggi E, Scarselli G, Romagnani S. Defective production of both leukemia inhibitory factor and type 2 T-helper cytokines by decidual T cells in unexplained recurrent abortions. Nat Med. (1998) 4:1020–4. 10.1038/2006
    1. Raghupathy R. Th1-type immunity is incompatible with successful pregnancy. Immunol Today. (1997) 18:478–82. 10.1016/S0167-5699(97)01127-4
    1. Saito S, Sakai M. Th1/Th2 balance in preeclampsia. J Reproduct Immunol. (2003) 59:161–73. 10.1016/S0165-0378(03)00045-7
    1. Chaouat G, Ledee-Bataille N, Zourbas S, Ostojic S, Dubanchet S, Martal J, et al. Cytokines, implantation and early abortion: re-examining the Th1/Th2 paradigm leads to question the single pathway, single therapy concept. Am J Reproduct Immunol. (2003) 50:177–86. 10.1034/j.1600-0897.2003.00080.x
    1. Bates MD, Quenby S, Takakuwa K, Johnson PM, Vince GS. Aberrant cytokine production by peripheral blood mononuclear cells in recurrent pregnancy loss? Hum Reprod. (2002) 17:2439–44. 10.1093/humrep/17.9.2439
    1. Chaouat G, Voisin GA. Regulatory T cell subpopulations in pregnancy. I. Evidence for suppressive activity of the early phase of MLR. J Immunol. (1979) 122:1383–8.
    1. Saito S, Nakashima A, Shima T, Ito M. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am J Reproduct Immunol. (2010) 63:601–10. 10.1111/j.1600-0897.2010.00852.x
    1. Jiang TT, Chaturvedi V, Ertelt JM, Kinder JM, Clark DR, Valent AM, et al. Regulatory T cells: new keys for further unlocking the enigma of fetal tolerance and pregnancy complications. J Immunol. (2014) 192:4949–56. 10.4049/jimmunol.1400498
    1. Wang WJ, Hao CF, Yi L, Yin GJ, Bao SH, Qiu LH, et al. Increased prevalence of T helper 17 (Th17) cells in peripheral blood and decidua in unexplained recurrent spontaneous abortion patients. J Reproduct Immunol. (2010) 84:164–70. 10.1016/j.jri.2009.12.003
    1. Azevedo RSS, De Sousa JR, Araujo MTF, Martins Filho AJ, De Alcantara BN, Araujo FMC, et al. In situ immune response and mechanisms of cell damage in central nervous system of fatal cases microcephaly by Zika virus. Sci Rep. (2018) 8:1. 10.1038/s41598-017-17765-5
    1. Piccinni MP, Lombardelli L, Logiodice F, Kullolli O, Parronchi P, Romagnani S. How pregnancy can affect autoimmune diseases progression? Clin Mol Allergy. (2016) 14:11. 10.1186/s12948-016-0048-x
    1. Logiodice F, Lombardelli L, Kullolli O, Haller H, Maggi E, Rukavina D, et al. Decidual Interleukin-22-Producing CD4+ T Cells (Th17/Th0/IL-22+ and Th17/Th2/IL-22+, Th2/IL-22+, Th0/IL-22+), Which Also Produce IL-4, Are Involved in the Success of Pregnancy. Int J Mol Sci. (2019) 20:428. 10.3390/ijms20020428
    1. Monteiro C, Kasahara TM, Castro JR, Sacramento PM, Hygino J, Centurião N, et al. Pregnancy favors the expansion of circulating functional follicular helper T Cells. J Reproduct Immunol. (2017) 121:1–10. 10.1016/j.jri.2017.04.007
    1. Tilburgs T, Meissner TB, Ferreira LMR, Mulder A, Musunuru K, Ye J, et al. NLRP2 is a suppressor of NF-kB signaling and HLA-C expression in human trophoblastsdagger, double dagger. Biol Reproduct. (2017) 96:831–42. 10.1093/biolre/iox009
    1. Billingham RE, Brent L, Medawar PB. ‘Actively acquired tolerance’ of foreign cells. 1953. J Immunol. (2010) 184:5–8. 10.4049/jimmunol.0990109
    1. Tersigni C, Redman CW, Dragovic R, Tannetta D, Scambia G, Di Simone N, et al. HLA-DR is aberrantly expressed at feto-maternal interface in pre-eclampsia. J Reproduct Immunol. (2018) 129:48–52. 10.1016/j.jri.2018.06.024
    1. Tilburgs T, Scherjon SA, Claas FH. Major histocompatibility complex (MHC)-mediated immune regulation of decidual leukocytes at the fetal-maternal interface. J Reproduct Immunol. (2010) 85:58–62. 10.1016/j.jri.2010.01.005
    1. Tilburgs T, Scherjon SA, van der Mast BJ, Haasnoot GW, Versteeg VDV-MM, Roelen DL, et al. Fetal-maternal HLA-C mismatch is associated with decidual T cell activation and induction of functional T regulatory cells. J Reproduct Immunol. (2009) 82:148–57. 10.1016/j.jri.2009.05.003
    1. Erlebacher A. Immunology of the maternal-fetal interface. Ann Rev Immunol. (2013) 31:387–411. 10.1146/annurev-immunol-032712-100003
    1. Erlebacher A, Vencato D, Price KA, Zhang D, Glimcher LH. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Investigat. (2007) 117:1399–411. 10.1172/JCI28214
    1. Papuchova H, Meissner TB, Li Q, Strominger JL, Tilburgs T. The dual role of HLA-C in tolerance and immunity at the maternal-fetal interface. Front Immunol. (2019) 10:2730. 10.3389/fimmu.2019.02730
    1. Mor G, Aldo P, Alvero AB. The unique immunological and microbial aspects of pregnancy. Nat Rev Immunol. (2017) 17:469–82. 10.1038/nri.2017.64
    1. Krey G, Frank P, Shaikly V, Barrientos G, Cordo-Russo R, Ringel F, et al. In vivo dendritic cell depletion reduces breeding efficiency, affecting implantation and early placental development in mice. J Mol Med (Berl). (2008) 86:999–1011. 10.1007/s00109-008-0379-2
    1. Germain SJ, Sacks GP, Sooranna SR, Sargent IL, Redman CW. Systemic inflammatory priming in normal pregnancy and preeclampsia: the role of circulating syncytiotrophoblast microparticles. J Immunol. (2007) 178:5949–56. 10.4049/jimmunol.178.9.5949
    1. Sargent IL, Borzychowski AM, Redman CW. NK cells and human pregnancy–an inflammatory view. Trends Immunol. (2006) 27:399–404. 10.1016/j.it.2006.06.009
    1. Gupta AK, Rusterholz C, Holzgreve W, Hahn S. Syncytiotrophoblast micro-particles do not induce apoptosis in peripheral T lymphocytes, but differ in their activity depending on the mode of preparation. J Reproduct Immunol. (2005) 68:15–26. 10.1016/j.jri.2005.05.003
    1. Knieke K, Lingel H, Chamaon K, Brunner-Weinzierl MC. Migration of Th1 lymphocytes is regulated by CD152 (CTLA-4)-mediated signaling via PI3 kinase-dependent Akt activation. PLoS One. (2012) 7:e31391. 10.1371/journal.pone.0031391
    1. Huang X, Cai Y, Ding M, Zheng B, Sun H, Zhou J. Human chorionic gonadotropin promotes recruitment of regulatory T cells in endometrium by inducing chemokine CCL2. J Reproduct Immunol. (2020) 137:102856. 10.1016/j.jri.2019.102856
    1. Tilburgs T, Roelen DL, van der Mast BJ, de Groot-Swings GM, Kleijburg C, Scherjon SA, et al. Evidence for a selective migration of fetus-specific CD4+CD25bright regulatory T cells from the peripheral blood to the decidua in human pregnancy. J Immunol. (2008) 180:5737–45. 10.4049/jimmunol.180.8.5737
    1. Saito S, Shiozaki A, Sasaki Y, Nakashima A, Shima T, Ito M. Regulatory T cells and regulatory natural killer (NK) cells play important roles in feto-maternal tolerance. Semin Immunopathol. (2007) 29:115–22. 10.1007/s00281-007-0067-2
    1. Saito S. Cytokine network at the feto-maternal interface. J Reproduct Immunol. (2000) 47:87–103. 10.1016/S0165-0378(00)00060-7
    1. Torchinsky A, Shepshelovich J, Orenstein H, Zaslavsky Z, Savion S, Carp H, et al. TNF-alpha protects embryos exposed to developmental toxicants. Am J Reproduct Immunol. (2003) 49:159–68. 10.1034/j.1600-0897.2003.01174.x
    1. Todt JC, Yang Y, Lei J, Lauria MR, Sorokin Y, Cotton DB, et al. Effects of tumor necrosis factor-alpha on human trophoblast cell adhesion and motility. Am J Reproduct Immunol. (1996) 36:65–71. 10.1111/j.1600-0897.1996.tb00141.x
    1. Bauer S, Pollheimer J, Hartmann J, Husslein P, Aplin JD, Knofler M. Tumor necrosis factor-alpha inhibits trophoblast migration through elevation of plasminogen activator inhibitor-1 in first-trimester villous explant cultures. J Clin Endocrinol Metabol. (2004) 89:812–22. 10.1210/jc.2003-031351
    1. Renaud SJ, Postovit LM, Macdonald-Goodfellow SK, McDonald GT, Caldwell JD, Graham CH. Activated macrophages inhibit human cytotrophoblast invasiveness in vitro. Biol Reproduct. (2005) 73:237–43. 10.1095/biolreprod.104.038000
    1. Seki H, Matuoka K, Inooku H, Takeda S. TNF-alpha from monocyte of patients with pre-eclampsia-induced apoptosis in human trophoblast cell line. J Obstetr Gynaecol Res. (2007) 33:408–16. 10.1111/j.1447-0756.2007.00551.x
    1. Hung TH, Charnock-Jones DS, Skepper JN, Burton GJ. Secretion of tumor necrosis factor-alpha from human placental tissues induced by hypoxia-reoxygenation causes endothelial cell activation in vitro: a potential mediator of the inflammatory response in preeclampsia. Am J Pathol. (2004) 164:1049–61. 10.1016/S0002-9440(10)63192-6
    1. Chen Q, Stone PR, McCowan LM, Chamley LW. Activated endothelial cells resist displacement by trophoblast in vitro. Placenta. (2007) 28:743–7. 10.1016/j.placenta.2006.10.004
    1. Meisser A, Chardonnens D, Campana A, Bischof P. Effects of tumour necrosis factor-alpha, interleukin-1 alpha, macrophage colony stimulating factor and transforming growth factor beta on trophoblastic matrix metalloproteinases. Mol Hum Reproduct. (1999) 5:252–60. 10.1093/molehr/5.3.252
    1. Lockwood CJ, Oner C, Uz YH, Kayisli UA, Huang SJ, Buchwalder LF, et al. Matrix metalloproteinase 9 (MMP9) expression in preeclamptic decidua and MMP9 induction by tumor necrosis factor alpha and interleukin 1 beta in human first trimester decidual cells. Biol Reproduct. (2008) 78:1064–72. 10.1095/biolreprod.107.063743
    1. Jokhi PP, King A, Sharkey AM, Smith SK, Loke YW. Screening for cytokine messenger ribonucleic acids in purified human decidual lymphocyte populations by the reverse-transcriptase polymerase chain reaction. J Immunol. (1994) 153:4427–35.
    1. Delassus S, Coutinho GC, Saucier C, Darche S, Kourilsky P. Differential cytokine expression in maternal blood and placenta during murine gestation. J Immunol. (1994) 152:2411–20.
    1. Ashkar AA, Di Santo JP, Croy BA. Interferon gamma contributes to initiation of uterine vascular modification, decidual integrity, and uterine natural killer cell maturation during normal murine pregnancy. J Exp Med. (2000) 192:259–70. 10.1084/jem.192.2.259
    1. Miyazaki S, Tsuda H, Sakai M, Hori S, Sasaki Y, Futatani T, et al. Predominance of Th2-promoting dendritic cells in early human pregnancy decidua. J Leukoc Biol. (2003) 74:514–22. 10.1189/jlb.1102566
    1. Ashkar AA, Croy BA. Interferon-gamma contributes to the normalcy of murine pregnancy. Biol Reproduct. (1999) 61:493–502. 10.1095/biolreprod61.2.493
    1. Raphael I, Nalawade S, Eagar TN, Forsthuber TG. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine. (2015) 74:5–17. 10.1016/j.cyto.2014.09.011
    1. Tayade C, Fang Y, Hilchie D, Croy BA. Lymphocyte contributions to altered endometrial angiogenesis during early and midgestation fetal loss. J Leukoc Biol. (2007) 82:877–86. 10.1189/jlb.0507330
    1. Murphy SP, Tayade C, Ashkar AA, Hatta K, Zhang J, Croy BA. Interferon gamma in successful pregnancies. Biol Reproduct. (2009) 80:848–59. 10.1095/biolreprod.108.073353
    1. Renaud SJ, Cotechini T, Quirt JS, Macdonald-Goodfellow SK, Othman M, Graham CH. Spontaneous pregnancy loss mediated by abnormal maternal inflammation in rats is linked to deficient uteroplacental perfusion. J Immunol. (2011) 186:1799–808. 10.4049/jimmunol.1002679
    1. Lin Y, Ren L, Wang W, Di J, Zeng S, Saito S. Effect of TLR3 and TLR7 activation in uterine NK cells from non-obese diabetic (n.d.) mice. J Reproduct Immunol. (2009) 82:12–23. 10.1016/j.jri.2009.03.004
    1. Ebina Y, Shimada S, Deguchi M, Maesawa Y, Iijima N, Yamada H. Divergence of helper, cytotoxic, and regulatory T cells in the decidua from miscarriage. Am J Reproduct Immunol. (2016) 76:199–204. 10.1111/aji.12546
    1. Jin LP, Fan DX, Zhang T, Guo PF, Li DJ. The costimulatory signal upregulation is associated with Th1 bias at the maternal-fetal interface in human miscarriage. Am J Reproduct Immunol. (2011) 66:270–8. 10.1111/j.1600-0897.2011.00997.x
    1. Makhseed M, Raghupathy R, Azizieh F, Omu A, Al-Shamali E, Ashkanani L. Th1 and Th2 cytokine profiles in recurrent aborters with successful pregnancy and with subsequent abortions. Hum Reprod. (2001) 16:2219–26. 10.1093/humrep/16.10.2219
    1. Kwak-Kim JY, Chung-Bang HS, Ng SC, Ntrivalas EI, Mangubat CP, Beaman KD, et al. Increased T helper 1 cytokine responses by circulating T cells are present in women with recurrent pregnancy losses and in infertile women with multiple implantation failures after IVF. Hum Reprod. (2003) 18:767–73. 10.1093/humrep/deg156
    1. Ng SC, Gilman-Sachs A, Thaker P, Beaman KD, Beer AE, Kwak-Kim J. Expression of intracellular Th1 and Th2 cytokines in women with recurrent spontaneous abortion, implantation failures after IVF/ET or normal pregnancy. Am J Reproduct Immunol. (2002) 48:77–86. 10.1034/j.1600-0897.2002.01105.x
    1. Ahmadi M, Abdolmohammadi-Vahid S, Ghaebi M, Aghebati-Maleki L, Afkham A, Danaii S, et al. Effect of Intravenous immunoglobulin on Th1 and Th2 lymphocytes and improvement of pregnancy outcome in recurrent pregnancy loss (RPL). Biomed Pharmacother Biomed Pharmacother. (2017) 92:1095–102. 10.1016/j.biopha.2017.06.001
    1. Muyayalo KP, Li ZH, Mor G, Liao AH. Modulatory effect of intravenous immunoglobulin on Th17/Treg cell balance in women with unexplained recurrent spontaneous abortion. Am J Reproduct Immunol. (2018) 80:e13018. 10.1111/aji.13018
    1. Nakagawa K, Kwak-Kim J, Kuroda K, Sugiyama R, Yamaguchi K. Immunosuppressive treatment using tacrolimus promotes pregnancy outcome in infertile women with repeated implantation failures. Am J Reproduct Immunol. (2017) 78:e12682. 10.1111/aji.12682
    1. Fu J, Li L, Qi L, Zhao L. A randomized controlled trial of etanercept in the treatment of refractory recurrent spontaneous abortion with innate immune disorders. Taiwan J Obstetr Gynecol. (2019) 58:621–5. 10.1016/j.tjog.2019.07.007
    1. Winger EE, Reed JL. Original article: treatment with tumor necrosis factor inhibitors and intravenous immunoglobulin improves live birth rates in women with recurrent spontaneous abortion. Am J Reproduct Immunol. (2008) 60:8–16. 10.1111/j.1600-0897.2008.00585.x
    1. Kim NY, Cho HJ, Kim HY, Yang KM, Ahn HK, Thornton S, et al. Thyroid autoimmunity and its association with cellular and humoral immunity in women with reproductive failures. Am J Reproduct Immunol. (2011) 65:78–87. 10.1111/j.1600-0897.2010.00911.x
    1. Guimarães PM, Scavuzzi BM, Stadtlober NP, Santos LFDRF, Lozovoy MAB, Iriyoda TMV, et al. Cytokines in systemic lupus erythematosus: far beyond Th1/Th2 dualism lupus: cytokine profiles. Immunol Cell Biol. (2017) 95:824–31. 10.1038/icb.2017.53
    1. Xiao J, Zhu F, Liu X, Xiong J. Th1/Th2/Th17/Treg expression in cultured PBMCs with antiphospholipid antibodies. Mol Med Rep. (2012) 6:1035–9. 10.3892/mmr.2012.1055
    1. Psianou K, Panagoulias I, Papanastasiou AD, de Lastic A-L, Rodi M, Spantidea PI, et al. Clinical and immunological parameters of Sjögren’s syndrome. Autoimmun Rev. (2018) 17:1053–64. 10.1016/j.autrev.2018.05.005
    1. Peña-Romero AG, García-Romero MT. Diagnosis and management of linear scleroderma in children. Curr Opin Pediatr. (2019) 31:482–90. 10.1097/MOP.0000000000000785
    1. Cellini M, Santaguida MG, Stramazzo I, Capriello S, Brusca N, Antonelli A, et al. Recurrent pregnancy loss in women with hashimoto’s thyroiditis with concurrent non-endocrine autoimmune disorders. Thyroid. (2020) 30:457–62. 10.1089/thy.2019.0456
    1. Kemp M, Thomas W. Antiphospholipid syndrome in obstetrics. Lupus. (2018) 27:28–31. 10.1177/0961203318801664
    1. Kwak-Kim J, Agcaoili MS, Aleta L, Liao A, Ota K, Dambaeva S, et al. Management of women with recurrent pregnancy losses and antiphospholipid antibody syndrome. Am J Reproduct Immunol. (2013) 69:596–607. 10.1111/aji.12114
    1. Kwak-Kim J, Lee SK, Gilman-Sachs A. Elevated Th1/Th2 cell ratios in a pregnant woman with a history of RSA, secondary Sjogren’s syndrome and rheumatoid arthritis complicated with one fetal demise of twin pregnancy. Am J Reproduct Immunol. (2007) 58:325–9. 10.1111/j.1600-0897.2007.00506.x
    1. Yi HJ, Kim JH, Koo HS, Bae JY, Cha SW, Yang KM. Elevated natural killer cell levels and autoimmunity synergistically decrease uterine blood flow during early pregnancy. Obstet Gynecol Sci. (2014) 57:208–15. 10.5468/ogs.2014.57.3.208
    1. Lin H, Mosmann TR, Guilbert L, Tuntipopipat S, Wegmann TG. Synthesis of T helper 2-type cytokines at the maternal-fetal interface. J Immunol. (1993) 151:4562–73.
    1. Daher S, de Arruda KGD, Blotta MH, Mamoni RL, Reck AP, Camano L, et al. Cytokines in recurrent pregnancy loss. J Reproduct Immunol. (2004) 62:151–7. 10.1016/j.jri.2003.10.004
    1. Fallon PG, Jolin HE, Smith P, Emson CL, Townsend MJ, Fallon R, et al. IL-4 induces characteristic Th2 responses even in the combined absence of IL-5, IL-9, and IL-13. Immunity. (2002) 17:7–17. 10.1016/S1074-7613(02)00332-1
    1. Piccinni MP, Scaletti C, Vultaggio A, Maggi E, Romagnani S. Defective production of LIF, M-CSF and Th2-type cytokines by T cells at fetomaternal interface is associated with pregnancy loss. J Reproduct Immunol. (2001) 52:35–43. 10.1016/S0165-0378(01)00111-5
    1. Piccinni MP, Lombardelli L, Logiodice F, Kullolli O, Romagnani S, Le Bouteiller P. T helper cell mediated-tolerance towards fetal allograft in successful pregnancy. Clin Mol Allergy. (2015) 13:9. 10.1186/s12948-015-0015-y
    1. Michimata T, Tsuda H, Sakai M, Fujimura M, Nagata K, Nakamura M, et al. Accumulation of CRTH2-positive T-helper 2 and T-cytotoxic 2 cells at implantation sites of human decidua in a prostaglandin D(2)-mediated manner. Mol Hum Reproduct. (2002) 8:181–7. 10.1093/molehr/8.2.181
    1. Tsuda H, Michimata T, Sakai M, Nagata K, Nakamura M, Saito S. A novel surface molecule of Th2- and Tc2-type cells, CRTH2 expression on human peripheral and decidual CD4+ and CD8+ T cells during the early stage of pregnancy. Clin Exp Immunol. (2001) 123:105–11. 10.1046/j.1365-2249.2001.01422.x
    1. Matthiesen L, Kalkunte S, Sharma S. Multiple pregnancy failures: an immunological paradigm. Am J Reproduct Immunol. (2012) 67:334–40. 10.1111/j.1600-0897.2012.01121.x
    1. Mitchell RE, Hassan M, Burton BR, Britton G, Hill EV, Verhagen J, et al. IL-4 enhances IL-10 production in Th1 cells: implications for Th1 and Th2 regulation. Sci Rep. (2017) 7:11315. 10.1038/s41598-017-11803-y
    1. Swain SL, Weinberg AD, English M, Huston G. IL-4 directs the development of Th2-like helper effectors. J Immunol. (1990) 145:3796–806.
    1. Holtan SG, Chen Y, Kaimal R, Creedon DJ, Enninga EA, Nevala WK, et al. Growth modeling of the maternal cytokine milieu throughout normal pregnancy: macrophage-derived chemokine decreases as inflammation/counterregulation increases. J Immunol Res. (2015) 2015:952571. 10.1155/2015/952571
    1. Illera VA, Perandones CE, Stunz LL, Mower DA, Jr., Ashman RF. Apoptosis in splenic B lymphocytes. Regulation by protein kinase C and IL-4. J Immunol. (1993) 151:2965–73.
    1. Jara LJ, Medina G, Cruz-Dominguez P, Navarro C, Vera-Lastra O, Saavedra MA. Risk factors of systemic lupus erythematosus flares during pregnancy. Immunol Res. (2014) 60:184–92. 10.1007/s12026-014-8577-1
    1. Breitfeld D, Ohl L, Kremmer E, Ellwart J, Sallusto F, Lipp M, et al. Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med. (2000) 192:1545–52. 10.1084/jem.192.11.1545
    1. Schaerli P, Willimann K, Lang AB, Lipp M, Loetscher P, Moser B. CXC chemokine receptor 5 expression defines follicular homing T cells with B cell helper function. J Exp Med. (2000) 192:1553–62. 10.1084/jem.192.11.1553
    1. Zeng W, Liu Z, Zhang S, Ren J, Ma X, Qin C, et al. Characterization of T follicular helper cells in allogeneic normal pregnancy and PDL1 blockage-induced abortion. Sci Rep. (2016) 6:36560–36560. 10.1038/srep36560
    1. Kaplan MH. Th9 cells: differentiation and disease. Immunol Rev. (2013) 252:104–15. 10.1111/imr.12028
    1. Micossé CL, von Meyenn OS, Kipfer E, Adam C, Simillion C, Jafari SMS, et al. Human “T(H)9” cells are a subpopulation of PPAR-γ(+) T(H)2 cells. Sci Immunol. (2019) 4:eaat5943. 10.1126/sciimmunol.aat5943
    1. Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, Buer J, et al. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat Immunol. (2008) 9:1341–6. 10.1038/ni.1659
    1. Putheti P, Awasthi A, Popoola J, Gao W, Strom TB. Human CD4+ memory T cells can become CD4+IL-9+ T cells. PLoS One. (2010) 5:e8706. 10.1371/journal.pone.0008706
    1. Demoulin JB, Renauld JC. Interleukin 9 and its receptor: an overview of structure and function. Int Rev Immunol. (1998) 16:345–64. 10.3109/08830189809043001
    1. Wilhelm C, Hirota K, Stieglitz B, Van Snick J, Tolaini M, Lahl K, et al. An IL-9 fate reporter demonstrates the induction of an innate IL-9 response in lung inflammation. Nat Immunol. (2011) 12:1071–7. 10.1038/ni.2133
    1. Nowak EC, Weaver CT, Turner H, Begum-Haque S, Becher B, Schreiner B, et al. IL-9 as a mediator of Th17-driven inflammatory disease. J Exp Med. (2009) 206:1653–60. 10.1084/jem.20090246
    1. Habbeddine M, Verbeke P, Karaz S, Bobé P, Kanellopoulos-Langevin C. Leukocyte population dynamics and detection of IL-9 as a major cytokine at the mouse fetal-maternal interface. PLoS One. (2014) 9:e107267. 10.1371/journal.pone.0107267
    1. Gomez-Lopez N, Olson DM, Robertson SA. Interleukin-6 controls uterine Th9 cells and CD8(+) T regulatory cells to accelerate parturition in mice. Immunol Cell Biol. (2016) 94:79–89. 10.1038/icb.2015.63
    1. Croy BA, Chantakru S, Esadeg S, Ashkar AA, Wei Q. Decidual natural killer cells: key regulators of placental development (a review). J Reproduct Immunol. (2002) 57:151–68. 10.1016/S0165-0378(02)00005-0
    1. Nakashima A, Ito M, Yoneda S, Shiozaki A, Hidaka T, Saito S. Circulating and decidual Th17 cell levels in healthy pregnancy. Am J Reproduct Immunol. (2010) 63:104–9. 10.1111/j.1600-0897.2009.00771.x
    1. Santner-Nanan B, Peek MJ, Khanam R, Richarts L, Zhu E, Fazekas B, et al. Systemic increase in the ratio between Foxp3+ and IL-17-producing CD4+ T cells in healthy pregnancy but not in preeclampsia. J Immunol. (2009) 183:7023–30. 10.4049/jimmunol.0901154
    1. Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. (2010) 10:479–89. 10.1038/nri2800
    1. Pongcharoen S, Supalap K. Interleukin-17 increased progesterone secretion by JEG-3 human choriocarcinoma cells. Am J Reproduct Immunol. (2009) 61:261–4. 10.1111/j.1600-0897.2009.00693.x
    1. Travis OK, White D, Pierce WA, Ge Y, Stubbs CY, Spradley FT, et al. Chronic infusion of interleukin-17 promotes hypertension, activation of cytolytic natural killer cells, and vascular dysfunction in pregnant rats. Physiol Rep. (2019) 7:e14038. 10.14814/phy2.14038
    1. Peck A, Mellins ED. Breaking old paradigms: Th17 cells in autoimmune arthritis. Clin Immunol. (2009) 132:295–304. 10.1016/j.clim.2009.03.522
    1. Crome SQ, Wang AY, Levings MK. Translational mini-review series on Th17 cells: function and regulation of human T helper 17 cells in health and disease. Clin Exp Immunol. (2010) 159:109–19. 10.1111/j.1365-2249.2009.04037.x
    1. Nakashima A, Ito M, Shima T, Bac ND, Hidaka T, Saito S. Accumulation of IL-17-positive cells in decidua of inevitable abortion cases. Am J Reproduct Immunol. (2010) 64:4–11. 10.1111/j.1600-0897.2010.00812.x
    1. Lee SK, Kim JY, Hur SE, Kim CJ, Na BJ, Lee M, et al. An imbalance in interleukin-17-producing T and Foxp3(+) regulatory T cells in women with idiopathic recurrent pregnancy loss. Hum Reprod. (2011) 26:2964–71. 10.1093/humrep/der301
    1. Lee SK, Kim JY, Lee M, Gilman-Sachs A, Kwak-Kim J. Th17 and regulatory T cells in women with recurrent pregnancy loss. Am J Reproduct Immunol. (2012) 67:311–8. 10.1111/j.1600-0897.2012.01116.x
    1. Wilczynski JR. Immunological analogy between allograft rejection, recurrent abortion and pre-eclampsia–the same basic mechanism? Hum Immunol. (2006) 67:492–511. 10.1016/j.humimm.2006.04.007
    1. Liu YS, Wu L, Tong XH, Wu LM, He GP, Zhou GX, et al. Study on the relationship between Th17 cells and unexplained recurrent spontaneous abortion. Am J Reproduct Immunol. (2011) 65:503–11. 10.1111/j.1600-0897.2010.00921.x
    1. Wu L, Li J, Xu HL, Xu B, Tong XH, Kwak-Kim J, et al. IL-7/IL-7R signaling pathway might play a role in recurrent pregnancy losses by increasing inflammatory Th17 cells and decreasing Treg cells. Am J Reproduct Immunol. (2016) 76:454–64. 10.1111/aji.12588
    1. Sereshki N, Gharagozloo M, Ostadi V, Ghahiri A, Roghaei MA, Mehrabian F, et al. Variations in T-helper 17 and regulatory T cells during the menstrual cycle in peripheral blood of women with recurrent spontaneous abortion. Int J Fertil Steril. (2014) 8:59–66.
    1. Jia L, Wu C. The biology and functions of Th22 cells. Adv Exp Med Biol. (2014) 841:209–30. 10.1007/978-94-017-9487-9_8
    1. Wang Y, Xu B, Li MQ, Li DJ, Jin LP. IL-22 secreted by decidual stromal cells and NK cells promotes the survival of human trophoblasts. Int J Clin Exp Pathol. (2013) 6:1781–90.
    1. Graham AC, Carr KD, Sieve AN, Indramohan M, Break TJ, Berg RE. IL-22 production is regulated by IL-23 during Listeria monocytogenes infection but is not required for bacterial clearance or tissue protection. PLoS One. (2011) 6:e17171. 10.1371/journal.pone.0017171
    1. Dambaeva S, Schneiderman S, Jaiswal MK, Agrawal V, Katara GK, Gilman-Sachs A, et al. Interleukin 22 prevents lipopolysaccharide- induced preterm labor in mice. Biol Reproduct. (2018) 98:299–308. 10.1093/biolre/iox182
    1. Plank MW, Kaiko GE, Maltby S, Weaver J, Tay HL, Shen W, et al. Th22 Cells form a distinct Th lineage from Th17 cells in vitro with unique transcriptional properties and tbet-dependent Th1 plasticity. J Immunol. (2017) 198:2182–90. 10.4049/jimmunol.1601480
    1. Roomandeh N, Saremi A, Arasteh J, Pak F, Mirmohammadkhani M, Kokhaei P, et al. Comparing serum levels of Th17 and treg cytokines in women with unexplained recurrent spontaneous abortion and fertile women. Iran J Immunol. (2018) 15:59–67.
    1. O’Hern Perfetto C, Fan X, Dahl S, Krieg S, Westphal LM, Bunker Lathi R, et al. Expression of interleukin-22 in decidua of patients with early pregnancy and unexplained recurrent pregnancy loss. J Assist Reprod Genet. (2015) 32:977–84. 10.1007/s10815-015-0481-7
    1. Lu KT, Kanno Y, Cannons JL, Handon R, Bible P, Elkahloun AG, et al. Functional and epigenetic studies reveal multistep differentiation and plasticity of in vitro-generated and in vivo-derived follicular T helper cells. Immunity. (2011) 35:622–32. 10.1016/j.immuni.2011.07.015
    1. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. (2000) 192:1027–34. 10.1084/jem.192.7.1027
    1. Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. (1996) 8:765–72. 10.1093/intimm/8.5.765
    1. Petroff MG, Chen L, Phillips TA, Azzola D, Sedlmayr P, Hunt JS. B7 family molecules are favorably positioned at the human maternal-fetal interface. Biol Reprod. (2003) 68:1496–504. 10.1095/biolreprod.102.010058
    1. Guleria I, Khosroshahi A, Ansari MJ, Habicht A, Azuma M, Yagita H, et al. A critical role for the programmed death ligand 1 in fetomaternal tolerance. J Exp Med. (2005) 202:231–7. 10.1084/jem.20050019
    1. Carter L, Fouser LA, Jussif J, Fitz L, Deng B, Wood CR, et al. PD-1:PD-L inhibitory pathway affects both CD4(+) and CD8(+) T cells and is overcome by IL-2. Eur J Immunol. (2002) 32:634–43. 10.1002/1521-4141(200203)32:3<634::AID-IMMU634>;2-9
    1. Zhang YH, Tian M, Tang MX, Liu ZZ, Liao AH. Recent insight into the role of the PD-1/PD-L1 pathway in feto-maternal tolerance and pregnancy. Am J Reprod Immunol. (2015) 74:201–8. 10.1111/aji.12365
    1. Fife BT, Guleria I, Bupp MG, Eagar TN, Tang Q, Bour-Jordan H, et al. Insulin-induced remission in new-onset NOD mice is maintained by the PD-1-PD-L1 pathway. J Exp Med. (2006) 203:2737–47. 10.1084/jem.20061577
    1. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. (2010) 236:219–42. 10.1111/j.1600-065X.2010.00923.x
    1. Asano T, Meguri Y, Yoshioka T, Kishi Y, Iwamoto M, Nakamura M, et al. PD-1 modulates regulatory T-cell homeostasis during low-dose interleukin-2 therapy. Blood. (2017) 129:2186–97. 10.1182/blood-2016-09-741629
    1. Wafula PO, Teles A, Schumacher A, Pohl K, Yagita H, Volk HD, et al. PD-1 but not CTLA-4 blockage abrogates the protective effect of regulatory T cells in a pregnancy murine model. Am J Reprod Immunol. (2009) 62:283–92. 10.1111/j.1600-0897.2009.00737.x
    1. Habicht A, Dada S, Jurewicz M, Fife BT, Yagita H, Azuma M, et al. A link between PDL1 and T regulatory cells in fetomaternal tolerance. J Immunol. (2007) 179:5211–9. 10.4049/jimmunol.179.8.5211
    1. Sugita S, Kawazoe Y, Imai A, Usui Y, Takahashi M, Mochizuki M. Suppression of IL-22-producing T helper 22 cells by RPE cells via PD-L1/PD-1 interactions. Invest Ophthalmol Vis Sci. (2013) 54:6926–33. 10.1167/iovs.13-12703
    1. Wang C, Lu Y, Chen L, Gao T, Yang Q, Zhu C, et al. Th9 cells are subjected to PD-1/PD-L1-mediated inhibition and are capable of promoting CD8 T cell expansion through IL-9R in colorectal cancer. Int Immunopharmacol. (2019) 78:106019. 10.1016/j.intimp.2019.106019
    1. Wang WJ, Garcia MDS, Deutsch G, Sung N, Yang X, He Q, et al. PD-1 and PD-L1 expression on T-cell subsets in women with unexplained recurrent pregnancy losses. Am J Reproduct Immunol. (2020) 83:e13230. 10.1111/aji.13230
    1. Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. (2005) 6:1245–52. 10.1038/ni1271
    1. Miyanishi N, Nishi N, Abe H, Kashio Y, Shinonaga R, Nakakita S, et al. Carbohydrate-recognition domains of galectin-9 are involved in intermolecular interaction with galectin-9 itself and other members of the galectin family. Glycobiology. (2007) 17:423–32. 10.1093/glycob/cwm001
    1. Seki M, Oomizu S, Sakata KM, Sakata A, Arikawa T, Watanabe K, et al. Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol. (2008) 127:78–88. 10.1016/j.clim.2008.01.006
    1. Oomizu S, Arikawa T, Niki T, Kadowaki T, Ueno M, Nishi N, et al. Cell surface galectin-9 expressing Th cells regulate Th17 and Foxp3+ Treg development by galectin-9 secretion. PLoS One. (2012) 7:e48574. 10.1371/journal.pone.0048574
    1. Zhao J, Lin B, Deng H, Zhi X, Li Y, Liu Y, et al. Decreased expression of TIM-3 on Th17 cells associated with ophthalmopathy in patients with graves’ disease. Curr Mol Med. (2018) 18:83–90. 10.2174/1566524018666180705105753
    1. Talukdar A, Sharma KA, Rai R, Deka D, Rao DN. Effect of coenzyme Q10 on Th1/Th2 paradigm in females with idiopathic recurrent pregnancy loss. Am J Reproduct Immunol. (2015) 74:169–80. 10.1111/aji.12376
    1. Wang S, Cao C, Piao H, Li Y, Tao Y, Zhang X, et al. Tim-3 protects decidual stromal cells from toll-like receptor-mediated apoptosis and inflammatory reactions and promotes Th2 bias at the maternal-fetal interface. Sci Rep. (2015) 5:9013. 10.1038/srep09013
    1. Wu M, Zhu Y, Zhao J, Ai H, Gong Q, Zhang J, et al. Soluble costimulatory molecule sTim3 regulates the differentiation of Th1 and Th2 in patients with unexplained recurrent spontaneous abortion. Int J Clin Exp Med. (2015) 8:8812–9.
    1. Zhuang X, Xia X, Liu L, Zhang Y, Zhang X, Wang C. Expression of Tim-3 in peripheral blood mononuclear cells and placental tissue in unexplained recurrent spontaneous abortion. Medicine (Baltimore). (2018) 97:e12099. 10.1097/MD.0000000000012099
    1. Li J, Li FF, Zuo W, Zhou Y, Hao HY, Dang J, et al. Up-regulated expression of Tim-3/Gal-9 at maternal-fetal interface in pregnant woman with recurrent spontaneous abortion. J Huazhong Univ Sci Technolog Med Sci. (2014) 34:586–90. 10.1007/s11596-014-1320-2
    1. Li F, Dang J, Jiang M, He M, Yang M, Li J, et al. Upregulation of Tim-3 expression at feto-maternal interface may explain embryo survival in the CBAxDBA/2 model of abortion. Am J Reproduct Immunol. (2018) 79:e12775. 10.1111/aji.12775
    1. Wang S, Li M, Sun F, Chen C, Ye J, Li D, et al. Altered frequency and function of spleen CTLA-4+Tim-3+ T cells are associated with miscarriage. Biol Reproduct. (2019) 19:ioz076. 10.1093/biolre/ioz076
    1. Hu XH, Tang MX, Mor G, Liao AH. Tim-3: Expression on immune cells and roles at the maternal-fetal interface. J Reprod Immunol. (2016) 118:92–9. 10.1016/j.jri.2016.10.113
    1. Li Y, Zhang J, Zhang D, Hong X, Tao Y, Wang S, et al. Tim-3 signaling in peripheral NK cells promotes maternal-fetal immune tolerance and alleviates pregnancy loss. Sci Signal. (2017) 10:eaah4323. 10.1126/scisignal.aah4323
    1. Wang S, Zhu X, Xu Y, Zhang D, Li Y, Tao Y, et al. Programmed cell death-1 (PD-1) and T-cell immunoglobulin mucin-3 (Tim-3) regulate CD4+ T cells to induce Type 2 helper T cell (Th2) bias at the maternal-fetal interface. Hum Reprod. (2016) 31:700–11. 10.1093/humrep/dew019
    1. Kao LC, Tulac S, Lobo S, Imani B, Yang JP, Germeyer A, et al. Global gene profiling in human endometrium during the window of implantation. Endocrinology. (2002) 143:2119–38. 10.1210/endo.143.6.8885
    1. Dambaeva S, Beaman K. Role of immunoregulatory cytokine IL-15 in the endometrium. In: Kwak-Kim J. editor. Endometrial Gene Expression. Cham: Springer; (2020). p. 66–74. 10.1007/978-3-030-28584-5_4
    1. Robertson SA, Care AS, Moldenhauer LM. Regulatory T cells in embryo implantation and the immune response to pregnancy. J Clin Investigat. (2018) 128:4224–35. 10.1172/JCI122182
    1. Wu L, Liao A, Gilman-Sachs A, Kwak-Kim J. T cell-related endometrial gene expression in normal and complicated pregnancies. In: Kwak-Kim J. editor. Endometrial Gene Expression: An Emerging Paradigm for Reproductive Disorders. Cham: Springer International Publishing; (2020). p. 51–66. 10.1007/978-3-030-28584-5_3
    1. Mekinian A, Cohen J, Alijotas-Reig J, Carbillon L, Nicaise-Roland P, Kayem G, et al. Unexplained recurrent miscarriage and recurrent implantation failure: is there a place for immunomodulation? Am J Reproduct Immunol. (2016) 76:8–28. 10.1111/aji.12493
    1. Wang WJ, Hao CF, Qu QL, Wang X, Qiu LH, Lin QD. The deregulation of regulatory T cells on interleukin-17-producing T helper cells in patients with unexplained early recurrent miscarriage. Hum Reprod. (2010) 25:2591–6. 10.1093/humrep/deq198
    1. Piccinni MP, Scaletti C, Maggi E, Romagnani S. Role of hormone-controlled Th1- and Th2-type cytokines in successful pregnancy. J Neuroimmunol. (2000) 109:30–3. 10.1016/S0165-5728(00)00299-X
    1. Piccinni MP, Giudizi MG, Biagiotti R, Beloni L, Giannarini L, Sampognaro S, et al. Progesterone favors the development of human T helper cells producing Th2-type cytokines and promotes both IL-4 production and membrane CD30 expression in established Th1 cell clones. J Immunol. (1995) 155:128–33.
    1. Petitbarat M, Serazin V, Dubanchet S, Wayner R, de Mazancourt P, Chaouat G, et al. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor inducible-14 might regulate the effects of interleukin 18 and 15 in the human endometrium. Fertil Steril. (2010) 94:1141–3. 10.1016/j.fertnstert.2009.10.049
    1. Fuertes MB, Molinero LL, Toscano MA, Ilarregui JM, Rubinstein N, Fainboim L, et al. Regulated expression of galectin-1 during T-cell activation involves Lck and Fyn kinases and signaling through MEK1/ERK, p38 MAP kinase and p70S6 kinase. Mol Cell Biochem. (2004) 267:177–85. 10.1023/B:MCBI.0000049376.50242.7f
    1. He M, Jiang M, Zhou Y, Li F, Yang M, Fan Y, et al. Impaired Gal-9 dysregulates the PBMC-induced Th1/Th2 imbalance in abortion-prone matings. J Immunol Res. (2018) 2018:9517842. 10.1155/2018/9517842
    1. Lombardelli L, Logiodice F, Aguerre-Girr M, Kullolli O, Haller H, Casart Y, et al. Interleukin-17-producing decidual CD4+ T cells are not deleterious for human pregnancy when they also produce interleukin-4. Clin Mol Allergy. (2016) 14:1. 10.1186/s12948-016-0039-y
    1. Raghupathy R. Pregnancy: success and failure within the Th1/Th2/Th3 paradigm. Semin Immunol. (2001) 13:219–27. 10.1006/smim.2001.0316
    1. Mor G, Cardenas I, Abrahams V, Guller S. Inflammation and pregnancy: the role of the immune system at the implantation site. Ann N Y Acad Sci. (2011) 1221:80–7. 10.1111/j.1749-6632.2010.05938.x
    1. Ali SB, Jeelall Y, Pennell CE, Hart R, McLean-Tooke A, Lucas M. The role of immunological testing and intervention in reproductive medicine: a fertile collaboration? Am J Reproduct Immunol. (2018) 79:e12784. 10.1111/aji.12784
    1. Lee YK, Turner H, Maynard CL, Oliver JR, Chen D, Elson CO, et al. Late developmental plasticity in the T helper 17 lineage. Immunity. (2009) 30:92–107. 10.1016/j.immuni.2008.11.005
    1. Cosmi L, Maggi L, Santarlasci V, Capone M, Cardilicchia E, Frosali F, et al. Identification of a novel subset of human circulating memory CD4(+) T cells that produce both IL-17A and IL-4. J Allergy Clin Immunol. (2010) 125:222–30. 10.1016/j.jaci.2009.10.012
    1. Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, Mazzinghi B, et al. Phenotypic and functional features of human Th17 cells. J Exp Med. (2007) 204:1849–61. 10.1084/jem.20070663
    1. Lee SK, Na BJ, Kim JY, Hur SE, Lee M, Gilman-Sachs A, et al. Determination of clinical cellular immune markers in women with recurrent pregnancy loss. Am J Reproduct Immunol. (2013) 70:398–411. 10.1111/aji.12137
    1. Salek Farrokhi A, Zarnani AH, Moazzeni SM. Mesenchymal stem cells therapy protects fetuses from resorption and induces Th2 type cytokines profile in abortion prone mouse model. Transpl Immunol. (2018) 47:26–31. 10.1016/j.trim.2018.01.002
    1. Carpentier PA, Dingman AL, Palmer TD. Placental TNF-α signaling in illness-induced complications of pregnancy. Am J Pathol. (2011) 178:2802–10. 10.1016/j.ajpath.2011.02.042
    1. Albaghdadi AJH, Kan FWK. Immunosuppression with tacrolimus improved implantation and rescued expression of uterine progesterone receptor and its co-regulators FKBP52 and PIASy at nidation in the obese and diabetic mice: Comparative studies with metformin. Mol Cell Endocrinol. (2018) 460:73–84. 10.1016/j.mce.2017.07.007
    1. Ota K, Dambaeva S, Han AR, Beaman K, Gilman-Sachs A, Kwak-Kim J. Vitamin D deficiency may be a risk factor for recurrent pregnancy losses by increasing cellular immunity and autoimmunity. Hum Reprod. (2014) 29:208–19. 10.1093/humrep/det424
    1. Ota K, Dambaeva S, Kim MW-I, Han A-R, Fukui A, Gilman-Sachs A, et al. 1,25-Dihydroxy-vitamin D3 regulates NK-cell cytotoxicity, cytokine secretion, and degranulation in women with recurrent pregnancy losses. Eur J Immunol. (2015) 45:3188–99. 10.1002/eji.201545541
    1. Wang SW, Zhong SY, Lou LJ, Hu ZF, Sun HY, Zhu HY. The effect of intravenous immunoglobulin passive immunotherapy on unexplained recurrent spontaneous abortion: a meta-analysis. Reprod Biomed Online. (2016) 33:720–36. 10.1016/j.rbmo.2016.08.025
    1. Ji J, Zhai H, Zhou H, Song S, Mor G, Liao A. The role and mechanism of vitamin D-mediated regulation of Treg/Th17 balance in recurrent pregnancy loss. Am J Reproduct Immunol. (2019) 81:e13112. 10.1111/aji.13112
    1. Ghasemnejad-berenji H, Ghaffari Novin M, Hajshafiha M, Nazarian H, Hashemi SM, Ilkhanizadeh B, et al. Immunomodulatory effects of hydroxychloroquine on Th1/Th2 balance in women with repeated implantation failure. Biomed Pharmacother. (2018) 107:1277–85. 10.1016/j.biopha.2018.08.027
    1. Ponticelli C, Moroni G. Fetal toxicity of immunosuppressive drugs in pregnancy. J Clin Med. (2018) 7:552. 10.3390/jcm7120552
    1. Kwak-Kim J, Ota K, Sung N, Huang C, Alsubki L, Lee S, et al. COVID-19 and immunomodulation treatment for women with reproductive failures. J Reproduct Immunol. (2020) 141:103168. 10.1016/j.jri.2020.103168
    1. Timilshina M, Kang Y, Dahal I, You Z, Nam TG, Kim KJ, et al. BJ-3105, a 6-alkoxypyridin-3-ol analog, impairs T cell differentiation and prevents experimental autoimmune encephalomyelitis disease progression. PLoS One. (2017) 12:e0168942. 10.1371/journal.pone.0168942
    1. Ahmad SF, Ansari MA, Nadeem A, Zoheir KMA, Bakheet SA, Alsaad AMS, et al. STA-21, a STAT-3 inhibitor, attenuates the development and progression of inflammation in collagen antibody-induced arthritis. Immunobiology. (2017) 222:206–17. 10.1016/j.imbio.2016.10.001
    1. Anthes E. Hard Labour: The Case for Testing Drugs on Pregnant Women. Oakville, ON: Mosaic Press; (2015).

Source: PubMed

3
Sottoscrivi