Regulatory T Cells Play a Role in a Subset of Idiopathic Preterm Labor/Birth and Adverse Neonatal Outcomes

Nardhy Gomez-Lopez, Marcia Arenas-Hernandez, Roberto Romero, Derek Miller, Valeria Garcia-Flores, Yaozhu Leng, Yi Xu, Jose Galaz, Sonia S Hassan, Chaur-Dong Hsu, Harley Tse, Carmen Sanchez-Torres, Bogdan Done, Adi L Tarca, Nardhy Gomez-Lopez, Marcia Arenas-Hernandez, Roberto Romero, Derek Miller, Valeria Garcia-Flores, Yaozhu Leng, Yi Xu, Jose Galaz, Sonia S Hassan, Chaur-Dong Hsu, Harley Tse, Carmen Sanchez-Torres, Bogdan Done, Adi L Tarca

Abstract

Regulatory T cells (Tregs) have been exhaustively investigated during early pregnancy; however, their role later in gestation is poorly understood. Herein, we report that functional Tregs are reduced at the maternal-fetal interface in a subset of women with idiopathic preterm labor/birth, which is accompanied by a concomitant increase in Tc17 cells. In mice, depletion of functional Tregs during late gestation induces preterm birth and adverse neonatal outcomes, which are rescued by the adoptive transfer of such cells. Treg depletion does not alter obstetrical parameters in the mother, yet it increases susceptibility to endotoxin-induced preterm birth. The mechanisms whereby depletion of Tregs induces adverse perinatal outcomes involve tissue-specific immune responses and mild systemic maternal inflammation, together with dysregulation of developmental and cellular processes in the placenta, in the absence of intra-amniotic inflammation. These findings provide mechanistic evidence supporting a role for Tregs in the pathophysiology of idiopathic preterm labor/birth and adverse neonatal outcomes.

Keywords: T cells; amniotic fluid; decidua; fetal growth restriction; maternal-fetal interface; myometrium; neonate; parturition; placenta; prematurity.

Conflict of interest statement

Declaration of Interests The authors declare no competing interests.

Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. Functional Tregs Are Reduced at…
Figure 1.. Functional Tregs Are Reduced at the Human Maternal-Fetal Interface in a Subset of Idiopathic PTL and birth
(A) Representative gating strategy used to sort Tregs and Teffs from the decidua. Tregs were co-cultured with Teffs, and Teff proliferation was measured by flow cytometry using CellTrace violet. (B) Representative plots showing the proliferation of Teffs, with the percentage of decidual Treg suppression of Teffs from preterm and term pregnancies. Suppression data are shown as means ± SEM. n = 6–8 per group. (C) Representative gating strategy used to identify Tregs in the decidua parietalis and decidua basalis. (D and E) Frequency of Tregs in the (D) decidua parietalis (n = 11–28 per group) or (E) decidua basalis (n = 13–28 per group) of women with PTNL, iPTL, iPTL+CI, or PTL+AI. (F and G) Frequency of Tregs in the (F) decidua parietalis (n = 13–19 per group) or (G) decidua basalis (n = 13–19 per group) of women with TNL, TIL, TIL+CI, or TIL+AI. Data are represented as medians with interquartile and minimum/maximum ranges. Statistical analysis was performed using the Mann-Whitney U-test. Demographic and clinical characteristics of the study population are shown in Tables S1 and S2.
Figure 2.. Tc17 Cells Are Increased at…
Figure 2.. Tc17 Cells Are Increased at the Human Maternal-Fetal Interface in Idiopathic PTL and birth
(A) Representative gating strategy used to identify Th17 cells and Tc17 cells in the decidua parietalis and decidua basalis. (B and C) Frequency of Th17 cells in the (B) decidua parietalis (n = 11–28 per group) or (C) decidua basalis (n = 13–28 per group) of women with PTNL, iPTL, iPTL+CI, or PTL+AI. (D and E) Frequency of Th17 cells in the (D) decidua parietalis (n = 13–19 per group) or (E) decidua basalis (n = 13–19 per group) of women with TNL, TIL, TIL+CI, or TIL+AI. (F and G) Frequency of Tc17 cells in the (F) decidua parietalis (n = 11–28 per group) or (G) decidua basalis (n = 13–28 per group) of women with PTNL, iPTL, iPTL+CI, or PTL+AI. (H and I) Frequency of Tc17 cells in the (H) decidua parietalis (n = 13–19 per group) or (I) decidua basalis (n = 13–19 per group) of women with TNL, TIL, TIL+CI, or TIL+AI. Data are represented as medians with interquartile and minimum/maximum ranges. Statistical analysis was performed using the Mann-Whitney U-test. Demographic and clinical characteristics of the study population are shown in Table S2.
Figure 3.. Depletion of Tregs Induces a…
Figure 3.. Depletion of Tregs Induces a Fraction of Preterm Births and Adverse Neonatal Outcomes
(A) Foxp3DTR dams underwent partial or total Treg depletion. Controls were injected with sterile 13 PBS. After the first pregnancy (P), a subset of Foxp3DTR dams underwent a second P and were again partially or totally Treg-depleted or were injected with sterile 13 PBS. (B) Frequencies of Tregs in the decidua, myometrium, peripheral blood, and placenta of partially or totally Treg-depleted Foxp3DTR dams (n = 5–7 per group). Data are represented as medians with interquartile and minimum/maximum ranges. (C) Preterm birth rates of non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (1st or 2nd P, n = 9–20 per group). Data are represented as means of percentages. (D) Percentage of survival from birth until 3 weeks postpartum for neonates born to non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (1st or 2nd P, n = 7–18 per group). (E–G) Weights of neonates born to non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams at weeks (E) 1, (F) 2, and (G) 3 postpartum (1st or 2nd P, n = 2–12 litters per group). Data are represented as violin plots with medians and minimum/maximum ranges. (H) Representative images of fetuses (and their placentas) from non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (n = 8–9 per group). (I and J) Weights of (I) fetuses and (J) their placentas from non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (n = 7 litters per group). Statistical analysis was performed using the Mantel-Cox test for survival curves, and Kruskal-Wallis or ANOVA tests with correction for multiple comparisons. See also Figures S1–S4.
Figure 4.. The Adoptive Transfer of Tregs…
Figure 4.. The Adoptive Transfer of Tregs Prevents Preterm Birth and Adverse Neonatal Outcomes
(A) Foxp3DTR dams underwent partial Treg depletion. On 14.5 and 16.5 dpc, Foxp3DTR dams received an adoptive transfer of Tregs from wild-type mice. (B) Preterm birth rates of partially Treg-depleted Foxp3DTR dams without or with the adoptive transfer of Tregs (n = 6–20 per group). Data are represented as means of percentages. (C) Percentage of survival from birth until 3 weeks postpartum for neonates born to partially Treg-depleted Foxp3DTR dams without or with the adoptive transfer of Tregs (n = 6–20 per group). (D) Foxp3DTR dams underwent partial Treg depletion. On 14.5 and 16.5 dpc, Foxp3DTR dams received an adoptive transfer of Tregs from EGFP mice. (E) Representative gating and histograms showing adoptively transferred GFP+ Tregs in the decidua, myometrium, placenta, and peripheral tissues (peripheral blood mononuclear cells [PBMCs], uterine-draining lymph nodes [ULNs], and spleen) of recipient Treg-depleted dams. Statistical analysis was performed using the Mantel-Cox test for survival curves.
Figure 5.. Maternal-Fetal Obstetrical Parameters upon Partial…
Figure 5.. Maternal-Fetal Obstetrical Parameters upon Partial or Total Treg Depletion
(A) Foxp3DTR dams underwent partial or total Treg depletion until 17.5 dpc on which body temperature, blood pressure, and Doppler determinations were performed (n = 8–9 per group). (B and C) Body temperature (B) and mean blood pressure (C) of non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (n = 8–9 per group). (D–F) Representative Doppler image of the uterine artery (D), which was used to determine (E) maternal heart rate, and (F) uterine artery pulsatility index of non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (n = 8–9 per group). (G–I) Representative Doppler image of the umbilical artery (G), which was used to determine (H) fetal heart rate, and (I) umbilical artery pulsatility index in fetuses of non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (n = 24–27 per group). (J) Representative images of the spleens and ULNs from non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams (n = 3 per group). (K) Representative images of neonates born to non-Treg-depleted-, partially Treg-depleted-, and totally Treg-depleted-Foxp3DTR dams on the day of birth (n = 3 per group). Red dotted squares indicate the presence of the milk band. Statistical analysis was performed using the Kruskal-Wallis or ANOVA tests with correction for multiple comparisons.
Figure 6.. Depletion of Tregs Induces a…
Figure 6.. Depletion of Tregs Induces a Mild Systemic Inflammatory Response in the Absence of Intra-Amniotic Inflammation
(A–H) Foxp3DTR dams underwent partial or total Treg depletion. Controls were injected with sterile 13 PBS. Mice were euthanized approximately 4 h after the second DT or PBS injection, and maternal plasma samples and amniotic fluid were collected. Concentrations of (A) IL-6, (B) CCL2, (C) IL-1β, (D) TNF-α, (E) IFNγ, (F) CCL7, (G) IL-22, and (H) IL-10 in the maternal plasma (n = 7 per group). Data are shown as medians with interquartile ranges and minimum/maximum ranges. C57BL/6 dams were intravenously injected with recombinant mouse IFNγ (1.4 pg/100 μL), CCL7 (218 pg/100 μL), IL-22 (48 pg/100 μL), or a combination of all three. Controls were injected with 100 μL of sterile 13 PBS alone. (I) Gestational length of cytokine-injected dams. Data are shown as means with standard deviations (n = 3–6 per group). (J) Rate of early-term or full-term delivery of cytokine-injected dams (n = 3–6 per group). (K) Percentage of survival from birth until 3 weeks postpartum for neonates born to cytokine-injected dams (n = 3–6 litters per group). (L–S) Concentrations of (L) IL-6, (M) CXCL10, (N) CCL2, (O) IL-1β, (P) TNF-α, (Q) IFNγ, (R) IL-10, and (S) IL-4 in the amniotic fluid (n = 5–7 per group). Data are shown as medians with interquartile ranges and minimum/maximum ranges. Statistical analysis was performed using the Mantel-Cox test for survival curves, and Kruskal-Wallis or ANOVA tests with correction for multiple comparisons. See also Figure S5.
Figure 7.. Depletion of Tregs Is Associated…
Figure 7.. Depletion of Tregs Is Associated with Altered Systemic and Local Cellular Immune Responses and Dysregulation of Developmental and Cellular Processes in the Placenta
Foxp3DTR dams underwent partial or total Treg depletion. Controls were injected with sterile 13 PBS. Mice were euthanized approximately 4 h after the second injection and the decidua, myometrium, placenta, and peripheral blood were collected for flow cytometry (all tissues, n = 5–7 per group) or for RNA-seq analysis (placenta only, n = 5 per group). (A) Heatmap visualization of changes in the log2-transformed frequencies of immune cell subsets in the decidua, myometrium, placenta, and peripheral blood of partially and totally Treg-depleted Foxp3DTR dams relative to controls. Red and green indicate increased and reduced abundance, respectively, relative to PBS controls. (B) Heatmap visualization of changes in gene expression in the placentas of partially and totally Treg-depleted Foxp3DTR dams and controls. Red indicates gene upregulation and green indicates gene downregulation relative to the average value in the PBS group. (C) Volcano plot showing genes differentially expressed between placentas from partially Treg-depleted Foxp3DTR dams and those from control dams. (D) Volcano plot showing genes differentially expressed between placental tissues from totally Treg-depleted Foxp3DTR dams and those from control dams. (E) Network of biological processes dysregulated in the placentas of totally Treg-depleted Foxp3DTR dams. Statistical analysis was performed using t-tests with false discovery rate adjustment. Asterisks indicate significant differences compared to controls after adjustment. See also Figure S6 and Tables S3, S4, S5, S6, S7, and S8.

References

    1. Aluvihare VR, Kallikourdis M, and Betz AG (2004). Regulatory T cells mediate maternal tolerance to the fetus. Nat. Immunol 5, 266–271.
    1. Anders S, McCarthy DJ, Chen Y, Okoniewski M, Smyth GK, Huber W, and Robinson MD (2013). Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat. Protoc 8, 1765–1786.
    1. Arck PC, and Hecher K (2013). Fetomaternal immune cross-talk and its consequences for maternal and offspring’s health. Nat. Med 19, 548–556.
    1. Arenas-Hernandez M, Sanchez-Rodriguez EN, Mial TN, Robertson SA, and Gomez-Lopez N (2015). Isolation of Leukocytes from the Murine Tissues at the Maternal-Fetal Interface. J. Vis. Exp 99, e52866.
    1. Arenas-Hernandez M, Romero R, St Louis D, Hassan SS, Kaye EB, and Gomez-Lopez N (2016). An imbalance between innate and adaptive immune cells at the maternal-fetal interface occurs prior to endotoxin-induced preterm birth. Cell. Mol. Immunol 13, 462–473.
    1. Arenas-Hernandez M, Romero R, Xu Y, Panaitescu B, Garcia-Flores V, Miller D, Ahn H, Done B, Hassan SS, Hsu CD, et al. (2019). Effector and Activated T Cells Induce Preterm Labor and Birth That Is Prevented by Treatment with Progesterone. J. Immunol 202, 2585–2608.
    1. Barros FC, Papageorghiou AT, Victora CG, Noble JA, Pang R, Iams J, Cheikh Ismail L, Goldenberg RL, Lambert A, Kramer MS, et al.; International Fetal and Newborn Growth Consortium for the 21st Century (2015). The distribution of clinical phenotypes of preterm birth syndrome: implications for prevention. JAMA Pediatr. 169, 220–229.
    1. Bazzoni F, Cassatella MA, Laudanna C, and Rossi F (1991). Phagocytosis of opsonized yeast induces tumor necrosis factor-alpha mRNA accumulation and protein release by human polymorphonuclear leukocytes. J. Leukoc. Biol 50, 223–228.
    1. Benjamini Y, and Hochberg Y (1995). Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300.
    1. Bersani I, De Carolis MP, Foell D, Weinhage T, Rossi ED, De Carolis S, Rubortone SA, Romagnoli C, and Speer CP (2015). Interleukin-22: biomarker of maternal and fetal inflammation? Immunol. Res 61, 4–10.
    1. Blencowe H, Cousens S, Oestergaard MZ, Chou D, Moller AB, Narwal R, Adler A, Vera Garcia C, Rohde S, Say L, and Lawn JE (2012). National, regional, and worldwide estimates of preterm birth rates in the year 2010 with time trends since 1990 for selected countries: a systematic analysis and implications. Lancet 379, 2162–2172.
    1. Bonney EA (2016). Immune Regulation in Pregnancy: A Matter of Perspective? Obstet. Gynecol. Clin. North Am 43, 679–698.
    1. Bonney EA, and Onyekwuluje J (2003). The H-Y response in mid-gestation and long after delivery in mice primed before pregnancy. Immunol. Invest 32, 71–81.
    1. Burton GJ, and Jauniaux E (2018). Pathophysiology of placental-derived fetal growth restriction. Am. J. Obstet. Gynecol 218 (2S), S745–S761.
    1. Care AS, Bourque SL, Morton JS, Hjartarson EP, Robertson SA, and Davidge ST (2018). Reduction in Regulatory T Cells in Early Pregnancy Causes Uterine Artery Dysfunction in Mice. Hypertension 72, 177–187.
    1. Chaouat G, Voisin GA, Escalier D, and Robert P (1979). Facilitation reaction (enhancing antibodies and suppressor cells) and rejection reaction (sensitized cells) from the mother to the paternal antigens of the conceptus. Clin. Exp. Immunol 35, 13–24.
    1. Chaouat G, Kolb JP, and Wegmann TG (1983). The murine placenta as an immunological barrier between the mother and the fetus. Immunol. Rev 75, 31–60.
    1. Chen T, Darrasse-Jèze G, Bergot AS, Courau T, Churlaud G, Valdivia K, Strominger JL, Ruocco MG, Chaouat G, and Klatzmann D (2013). Self-specific memory regulatory T cells protect embryos at implantation in mice. J. Immunol 191, 2273–2281.
    1. Cobo T, Tsiartas P, Kacerovsky M, Holst RM, Hougaard DM, Skogstrand K, Wennerholm UB, Hagberg H, and Jacobsson B (2013). Maternal inflammatory response to microbial invasion of the amniotic cavity: analyses of multiple proteins in the maternal serum. Acta Obstet. Gynecol. Scand 92, 61–68.
    1. Combs CA, Gravett M, Garite TJ, Hickok DE, Lapidus J, Porreco R, Rael J, Grove T, Morgan TK, Clewell W, et al.; ProteoGenix/Obstetrix Collaborative Research Network (2014). Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am. J. Obstet. Gynecol 210, 125.e1–125.e15.
    1. Couper KN, Blount DG, de Souza JB, Suffia I, Belkaid Y, and Riley EM (2007). Incomplete depletion and rapid regeneration of Foxp3+ regulatory T cells following anti-CD25 treatment in malaria-infected mice. J. Immunol 178, 4136–4146.
    1. Cox SM, Casey ML, and MacDonald PC (1997). Accumulation of interleukin-1beta and interleukin-6 in amniotic fluid: a sequela of labour at term and preterm. Hum. Reprod. Update 3, 517–527.
    1. Cruciani L, Romero R, Vaisbuch E, Kusanovic JP, Chaiworapongsa T, Mazaki-Tovi S, Dong Z, Kim SK, Ogge G, Yeo L, et al. (2010). Pentraxin 3 in maternal circulation: an association with preterm labor and preterm PROM, but not with intra-amniotic infection/inflammation. J. Matern. Fetal Neonatal Med 23, 1097–1105.
    1. D’Andrea A, Aste-Amezaga M, Valiante NM, Ma X, Kubin M, and Trinchieri G (1993). Interleukin 10 (IL-10) inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells. J. Exp. Med 178, 1041–1048.
    1. Darrasse-Jèze G, Klatzmann D, Charlotte F, Salomon BL, and Cohen JL (2006). CD4+CD25+ regulatory/suppressor T cells prevent allogeneic fetus rejection in mice. Immunol. Lett 102, 106–109.
    1. de Waal Malefyt R, Yssel H, and de Vries JE (1993). Direct effects of IL-10 on subsets of human CD4+ T cell clones and resting T cells. Specific inhibition of IL-2 production and proliferation. J. Immunol 150, 4754–4765.
    1. Deng W, Yuan J, Cha J, Sun X, Bartos A, Yagita H, Hirota Y, and Dey SK (2019). Endothelial Cells in the Decidual Bed Are Potential Therapeutic Targets for Preterm Birth Prevention. Cell Rep. 27, 1755–1768.
    1. Doisne JM, Balmas E, Boulenouar S, Gaynor LM, Kieckbusch J, Gardner L, Hawkes DA, Barbara CF, Sharkey AM, Brady HJ, et al. (2015). Composition, Development, and Function of Uterine Innate Lymphoid Cells. J. Immunol 195, 3937–3945.
    1. Doncheva NT, Morris JH, Gorodkin J, and Jensen LJ (2019). Cytoscape StringApp: Network Analysis and Visualization of Proteomics Data. J. Proteome Res 18, 623–632.
    1. Dudley DJ, Hunter C, Mitchell MD, and Varner MW (1997). Amniotic fluid interleukin-10 (IL-10) concentrations during pregnancy and with labor. J. Reprod. Immunol 33, 147–156.
    1. Elovitz MA, Gajer P, Riis V, Brown AG, Humphrys MS, Holm JB, and Ravel J (2019). Cervicovaginal microbiota and local immune response modulate the risk of spontaneous preterm delivery. Nat. Commun 10, 1305.
    1. Erlebacher A (2013). Immunology of the maternal-fetal interface. Annu. Rev. Immunol 31, 387–411.
    1. Esplin MS, Romero R, Chaiworapongsa T, Kim YM, Edwin S, Gomez R, Mazor M, and Adashi EY (2005). Monocyte chemotactic protein-1 is increased in the amniotic fluid of women who deliver preterm in the presence or absence of intra-amniotic infection. J. Matern. Fetal Neonatal Med 17, 365–373.
    1. Fan MY, Low JS, Tanimine N, Finn KK, Priyadharshini B, Germana SK, Kaech SM, and Turka LA (2018). Differential Roles of IL-2 Signaling in Developing versus Mature Tregs. Cell Rep. 25, 1204–1213.
    1. Fenton MJ, Buras JA, and Donnelly RP (1992). IL-4 reciprocally regulates IL-1 and IL-1 receptor antagonist expression in human monocytes. J. Immunol 149, 1283–1288.
    1. Fettweis JM, Serrano MG, Brooks JP, Edwards DJ, Girerd PH, Parikh HI, Huang B, Arodz TJ, Edupuganti L, Glascock AL, et al. (2019). The vaginal microbiome and preterm birth. Nat. Med 25, 1012–1021.
    1. Figueiredo AS, and Schumacher A (2016). The T helper type 17/regulatory T cell paradigm in pregnancy. Immunology 148, 13–21.
    1. Fossiez F, Djossou O, Chomarat P, Flores-Romo L, Ait-Yahia S, Maat C, Pin JJ, Garrone P, Garcia E, Saeland S, et al. (1996). T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J. Exp. Med 183, 2593–2603.
    1. Fu B, Zhou Y, Ni X, Tong X, Xu X, Dong Z, Sun R, Tian Z, and Wei H (2017). Natural Killer Cells Promote Fetal Development through the Secretion of Growth-Promoting Factors. Immunity 47, 1100–1113.
    1. Garcia-Flores V, Romero R, Miller D, Xu Y, Done B, Veerapaneni C, Leng Y, Arenas-Hernandez M, Khan N, Panaitescu B, et al. (2018). Inflammation-Induced Adverse Pregnancy and Neonatal Outcomes Can Be Improved by the Immunomodulatory Peptide Exendin-4. Front. Immunol 9, 1291.
    1. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al. (2004). Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80.
    1. Gervasi MT, Chaiworapongsa T, Naccasha N, Blackwell S, Yoon BH, Maymon E, and Romero R (2001). Phenotypic and metabolic characteristics of maternal monocytes and granulocytes in preterm labor with intact membranes. Am. J. Obstet. Gynecol 185, 1124–1129.
    1. Gervasi MT, Romero R, Bracalente G, Erez O, Dong Z, Hassan SS, Yeo L, Yoon BH, and Chaiworapongsa T (2012). Midtrimester amniotic fluid concentrations of interleukin-6 and interferon-gamma-inducible protein-10: evidence for heterogeneity of intra-amniotic inflammation and associations with spontaneous early (<32 weeks) and late (>32 weeks) preterm delivery. J. Perinat. Med 40, 329–343.
    1. Gill N, Leng Y, Romero R, Xu Y, Panaitescu B, Miller D, Arif A, Mumuni S, Qureshi F, Hsu CD, et al. (2019). The immunophenotype of decidual macrophages in acute atherosis. Am. J. Reprod. Immunol 81, e13098.
    1. Goldenberg RL, Culhane JF, Iams JD, and Romero R (2008). Epidemiology and causes of preterm birth. Lancet 371, 75–84.
    1. Gomez-Lopez N, and Laresgoiti-Servitje E (2012). T regulatory cells: regulating both term and preterm labor? Immunol. Cell Biol 90, 919–920.
    1. Gomez-Lopez N, Vega-Sanchez R, Castillo-Castrejon M, Romero R, Cubeiro-Arreola K, and Vadillo-Ortega F (2013). Evidence for a role for the adaptive immune response in human term parturition. Am. J. Reprod. Immunol 69, 212–230.
    1. Gomez-Lopez N, StLouis D, Lehr MA, Sanchez-Rodriguez EN, and Arenas-Hernandez M (2014). Immune cells in term and preterm labor. Cell. Mol. Immunol 11, 571–581.
    1. Gomez-Lopez N, Romero R, Arenas-Hernandez M, Ahn H, Panaitescu B, Vadillo-Ortega F, Sanchez-Torres C, Salisbury KS, and Hassan SS (2016a). In vivo T-cell activation by a monoclonal aCD3ε antibody induces preterm labor and birth. Am. J. Reprod. Immunol 76, 386–390.
    1. Gomez-Lopez N, Romero R, Plazyo O, Panaitescu B, Furcron AE, Miller D, Roumayah T, Flom E, and Hassan SS (2016b). Intra-Amniotic Administration of HMGB1 Induces Spontaneous Preterm Labor and Birth. Am. J. Reprod. Immunol 75, 3–7.
    1. Gomez-Lopez N, Romero R, Arenas-Hernandez M, Schwenkel G, St Louis D, Hassan SS, and Mial TN (2017a). In vivo activation of invariant natural killer T cells induces systemic and local alterations in T-cell subsets prior to preterm birth. Clin. Exp. Immunol 189, 211–225.
    1. Gomez-Lopez N, Romero R, Plazyo O, Schwenkel G, Garcia-Flores V, Unkel R, Xu Y, Leng Y, Hassan SS, Panaitescu B, et al. (2017b). Preterm labor in the absence of acute histologic chorioamnionitis is characterized by cellular senescence of the chorioamniotic membranes. Am. J. Obstet. Gynecol 217, 592.e1–592.e17.
    1. Gomez-Lopez N, Romero R, Arenas-Hernandez M, Panaitescu B, Garcia-Flores V, Mial TN, Sahi A, and Hassan SS (2018). Intra-amniotic administration of lipopolysaccharide induces spontaneous preterm labor and birth in the absence of a body temperature change. J. Matern. Fetal Neonatal Med 31, 439–446.
    1. Gomez-Lopez N, Romero R, Garcia-Flores V, Leng Y, Miller D, Hassan SS, Hsu CD, and Panaitescu B (2019). Inhibition of the NLRP3 inflammasome can prevent sterile intra-amniotic inflammation, preterm labor/birth, and adverse neonatal outcomes. Biol. Reprod 100, 1306–1318.
    1. Gousopoulos E, Proulx ST, Bachmann SB, Scholl J, Dionyssiou D, Demiri E, Halin C, Dieterich LC, and Detmar M (2016). Regulatory T cell transfer ameliorates lymphedema and promotes lymphatic vessel function. JCI Insight 1, e89081.
    1. Gravett MG, Witkin SS, Haluska GJ, Edwards JL, Cook MJ, and Novy MJ (1994). An experimental model for intraamniotic infection and preterm labor in rhesus monkeys. Am. J. Obstet. Gynecol 171, 1660–1667.
    1. Gustafsson C, Mjösberg J, Matussek A, Geffers R, Matthiesen L, Berg G, Sharma S, Buer J, and Ernerudh J (2008). Gene expression profiling of human decidual macrophages: evidence for immunosuppressive phenotype. PLoS ONE 3, e2078.
    1. Hamada H, Garcia-Hernandez Mde.L., Reome JB, Misra SK, Strutt TM, McKinstry KK, Cooper AM, Swain SL, and Dutton RW (2009). Tc17, a unique subset of CD8 T cells that can protect against lethal influenza challenge. J. Immunol 182, 3469–3481.
    1. Hart PH, Vitti GF, Burgess DR, Whitty GA, Piccoli DS, and Hamilton JA (1989). Potential antiinflammatory effects of interleukin 4: suppression of human monocyte tumor necrosis factor alpha, interleukin 1, and prostaglandin E2. Proc. Natl. Acad. Sci. USA 86, 3803–3807.
    1. Heikkinen J, Möttönen M, Alanen A, and Lassila O (2004). Phenotypic characterization of regulatory T cells in the human decidua. Clin. Exp. Immunol 136, 373–378.
    1. Holt R, Timmons BC, Akgul Y, Akins ML, and Mahendroo M (2011). The molecular mechanisms of cervical ripening differ between term and preterm birth. Endocrinology 152, 1036–1046.
    1. Houser BL, Tilburgs T, Hill J, Nicotra ML, and Strominger JL (2011). Two unique human decidual macrophage populations. J. Immunol 186, 2633–2642.
    1. Howson CP, Kinney MV, McDougall L, and Lawn JE; Born Too Soon Preterm Birth Action Group (2013). Born too soon: preterm birth matters. Reprod. Health 10 (Suppl 1), S1.
    1. Huber M, Heink S, Grothe H, Guralnik A, Reinhard K, Elflein K, Hünig T, Mittrücker HW, Brüstle A, Kamradt T, and Lohoff M (2009). A Th17-like developmental process leads to CD8(+) Tc17 cells with reduced cytotoxic activity. Eur. J. Immunol 39, 1716–1725.
    1. Hunt JS, Manning LS, and Wood GW (1984). Macrophages in murine uterus are immunosuppressive. Cell. Immunol 85, 499–510.
    1. Inada K, Shima T, Nakashima A, Aoki K, Ito M, and Saito S (2013). Characterization of regulatory T cells in decidua of miscarriage cases with abnormal or normal fetal chromosomal content. J. Reprod. Immunol 97, 104–111.
    1. Kahn DA, and Baltimore D (2010). Pregnancy induces a fetal antigen-specific maternal T regulatory cell response that contributes to tolerance. Proc. Natl. Acad. Sci. USA 107, 9299–9304.
    1. Kemper C, Chan AC, Green JM, Brett KA, Murphy KM, and Atkinson JP (2003). Activation of human CD4+ cells with CD3 and CD46 induces a T-regulatory cell 1 phenotype. Nature 421, 388–392.
    1. Kieckbusch J, Balmas E, Hawkes DA, and Colucci F (2015). Disrupted PI3K p110d Signaling Dysregulates Maternal Immune Cells and Increases Fetal Mortality In Mice. Cell Rep. 13, 2817–2828.
    1. Kim JM, Rasmussen JP, and Rudensky AY (2007). Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol 8, 191–197.
    1. Kim MJ, Romero R, Kim CJ, Tarca AL, Chhauy S, LaJeunesse C, Lee DC, Draghici S, Gotsch F, Kusanovic JP, et al. (2009). Villitis of unknown etiology is associated with a distinct pattern of chemokine up-regulation in the feto-maternal and placental compartments: implications for conjoint maternal allograft rejection and maternal anti-fetal graft-versus-host disease. J. Immunol 182, 3919–3927.
    1. Kim CJ, Romero R, Kusanovic JP, Yoo W, Dong Z, Topping V, Gotsch F, Yoon BH, Chi JG, and Kim JS (2010). The frequency, clinical significance, and pathological features of chronic chorioamnionitis: a lesion associated with spontaneous preterm birth. Mod. Pathol 23, 1000–1011.
    1. Kim CJ, Romero R, Chaemsaithong P, Chaiyasit N, Yoon BH, and Kim YM (2015a). Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am. J. Obstet. Gynecol 213 (Suppl 4), S29–S52.
    1. Kim CJ, Romero R, Chaemsaithong P, and Kim JS (2015b). Chronic inflammation of the placenta: definition, classification, pathogenesis, and clinical significance. Am. J. Obstet. Gynecol 213 (Suppl 4), S53–S69.
    1. Kohm AP, McMahon JS, Podojil JR, Begolka WS, DeGutes M, Kasprowicz DJ, Ziegler SF, and Miller SD (2006). Cutting Edge: Anti-CD25 monoclonal antibody injection results in the functional inactivation, not depletion, of CD4+CD25+ T regulatory cells. J. Immunol 176, 3301–3305.
    1. Laudanski P, Lemancewicz A, Kuc P, Charkiewicz K, Ramotowska B, Kretowska M, Jasinska E, Raba G, Karwasik-Kajszczarek K, Kraczkowski J, and Laudanski T (2014). Chemokines profiling of patients with preterm birth. Mediators Inflamm. 2014, 185758.
    1. Lee J, Romero R, Xu Y, Kim JS, Topping V, Yoo W, Kusanovic JP, Chaiworapongsa T, Hassan SS, Yoon BH, and Kim CJ (2011). A signature of maternal anti-fetal rejection in spontaneous preterm birth: chronic chorioamnionitis, anti-human leukocyte antigen antibodies, and C4d. PLoS ONE 6, e16806.
    1. Leng Y, Romero R, Xu Y, Galaz J, Slutsky R, Arenas-Hernandez M, Garcia-Flores V, Motomura K, Hassan SS, Reboldi A, and Gomez-Lopez N (2019). Are B cells altered in the decidua of women with preterm or term labor? Am. J. Reprod. Immunol 81, e13102.
    1. Li Y, Zhang J, Zhang D, Hong X, Tao Y, Wang S, Xu Y, Piao H, Yin W, Yu M, et al. (2017). Tim-3 signaling in peripheral NK cells promotes maternal-fetal immune tolerance and alleviates pregnancy loss. Sci. Signal 10, eaah4323.
    1. Liu L, Oza S, Hogan D, Perin J, Rudan I, Lawn JE, Cousens S, Mathers C, and Black RE (2015). Global, regional, and national causes of child mortality in 2000–13, with projections to inform post-2015 priorities: an updated systematic analysis. Lancet 385, 430–440.
    1. Loewendorf AI, Nguyen TA, Yesayan MN, and Kahn DA (2015). Pre-eclampsia is Characterized by Fetal NK Cell Activation and a Reduction in Regulatory T Cells. Am. J. Reprod. Immunol 74, 258–267.
    1. McGeachy MJ, Cua DJ, and Gaffen SL (2019). The IL-17 Family of Cytokines in Health and Disease. Immunity 50, 892–906.
    1. Menard LC, Minns LA, Darche S, Mielcarz DW, Foureau DM, Roos D, Dzierszinski F, Kasper LH, and Buzoni-Gatel D (2007). B cells amplify IFN-gamma production by T cells via a TNF-alpha-mediated mechanism. J. Immunol 179, 4857–4866.
    1. Miller D, Motomura K, Garcia-Flores V, Romero R, and Gomez-Lopez N (2018). Innate Lymphoid Cells in the Maternal and Fetal Compartments. Front. Immunol 9, 2396.
    1. Mlecnik B, Galon J, and Bindea G (2019). Automated exploration of gene ontology term and pathway networks with ClueGO-REST. Bioinformatics 35, 3864–3866.
    1. Mukhopadhyay D, Weaver L, Tobin R, Henderson S, Beeram M, Newell-Rogers MK, and Perger L (2014). Intrauterine growth restriction and prematurity influence regulatory T cell development in newborns. J. Pediatr. Surg 49, 727–732.
    1. Munder M, Schneider H, Luckner C, Giese T, Langhans CD, Fuentes JM, Kropf P, Mueller I, Kolb A, Modolell M, and Ho AD (2006). Suppression of T-cell functions by human granulocyte arginase. Blood 108, 1627–1634.
    1. Nguyen TA, Kahn DA, and Loewendorf AI (2017). Maternal-Fetal rejection reactions are unconstrained in preeclamptic women. PLoS ONE 12, e0188250.
    1. Nguyen TA, Kahn DA, and Loewendorf AI (2018). Placental implantation over prior cesarean scar causes activation of fetal regulatory T cells. Immun. Inflamm. Dis 6, 256–263.
    1. Oh KJ, Kim SM, Hong JS, Maymon E, Erez O, Panaitescu B, Gomez-Lopez N, Romero R, and Yoon BH (2017). Twenty-four percent of patients with clinical chorioamnionitis in preterm gestations have no evidence of either culture-proven intraamniotic infection or intraamniotic inflammation. Am. J. Obstet. Gynecol 216, 604.e1–604.e11.
    1. Onishi RM, and Gaffen SL (2010). Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology 129, 311–321.
    1. Otasek D, Morris JH, Bouças J, Pico AR, and Demchak B (2019). Cytoscape Automation: empowering workflow-based network analysis. Genome Biol. 20, 185.
    1. Paquette AG, Brockway HM, Price ND, and Muglia LJ (2018). Comparative transcriptomic analysis of human placentae at term and preterm delivery. Biol. Reprod 98, 89–101.
    1. Park H, Park KH, Kim YM, Kook SY, Jeon SJ, and Yoo HN (2018). Plasma inflammatory and immune proteins as predictors of intra-amniotic infection and spontaneous preterm delivery in women with preterm labor: a retrospective study. BMC Pregnancy Childbirth 18, 146.
    1. Patro R, Duggal G, Love MI, Irizarry RA, and Kingsford C (2017). Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419.
    1. Pereyra S, Sosa C, Bertoni B, and Sapiro R (2019). Transcriptomic analysis of fetal membranes reveals pathways involved in preterm birth. BMC Med. Genomics 12, 53.
    1. Petroff MG (2005). Immune interactions at the maternal-fetal interface. J. Reprod. Immunol 68, 1–13.
    1. Pique-Regi R, Romero R, Tarca AL, Sendler ED, Xu Y, Garcia-Flores V, Leng Y, Luca F, Hassan SS, and Gomez-Lopez N (2019). Single cell transcriptional signatures of the human placenta in term and preterm parturition. eLife 8, e52004.
    1. Polanczyk MJ, Hopke C, Huan J, Vandenbark AA, and Offner H (2005). Enhanced FoxP3 expression and Treg cell function in pregnant and estrogen-treated mice. J. Neuroimmunol 170, 85–92.
    1. Poon LC, Shennan A, Hyett JA, Kapur A, Hadar E, Divakar H, McAu-liffe F, da Silva Costa F, von Dadelszen P, McIntyre HD, et al. (2019). The International Federation of Gynecology and Obstetrics (FIGO) initiative on pre-eclampsia: A pragmatic guide for first-trimester screening and prevention. Int. J. Gynaecol. Obstet 145 (Suppl 1), 1–33.
    1. PrabhuDas M, Bonney E, Caron K, Dey S, Erlebacher A, Fazleabas A, Fisher S, Golos T, Matzuk M, McCune JM, et al. (2015). Immune mechanisms at the maternal-fetal interface: perspectives and challenges. Nat. Immunol 16, 328–334.
    1. Redline RW (2006). Inflammatory responses in the placenta and umbilical cord. Semin. Fetal Neonatal Med 11, 296–301.
    1. Redline RW, and Patterson P (1994). Patterns of placental injury. Correlations with gestational age, placental weight, and clinical diagnoses. Arch. Pathol. Lab. Med 118, 698–701.
    1. Redline RW, Faye-Petersen O, Heller D, Qureshi F, Savell V, and Vogler C; Society for Pediatric Pathology, Perinatal Section, Amniotic Fluid Infection Nosology Committee (2003). Amniotic infection syndrome: nosology and reproducibility of placental reaction patterns. Pediatr. Dev. Pathol 6, 435–448.
    1. Rinaldi SF, Makieva S, Saunders PT, Rossi AG, and Norman JE (2017). Immune cell and transcriptomic analysis of the human decidua in term and preterm parturition. Mol. Hum. Reprod 23, 708–724.
    1. Robertson SA, Guerin LR, Moldenhauer LM, and Hayball JD (2009). Activating T regulatory cells for tolerance in early pregnancy—the contribution of seminal fluid. J. Reprod. Immunol 83, 109–116.
    1. Robertson SA, Care AS, and Moldenhauer LM (2018). Regulatory T cells in embryo implantation and the immune response to pregnancy. J. Clin. Invest 128, 4224–4235.
    1. Romero R, Mazor M, Wu YK, Sirtori M, Oyarzun E, Mitchell MD, and Hobbins JC (1988). Infection in the pathogenesis of preterm labor. Semin. Perinatol 12, 262–279.
    1. Romero R, Sirtori M, Oyarzun E, Avila C, Mazor M, Callahan R, Sabo V, Athanassiadis AP, and Hobbins JC (1989). Infection and labor. V. Prevalence, microbiology, and clinical significance of intraamniotic infection in women with preterm labor and intact membranes. Am. J. Obstet. Gynecol 161, 817–824.
    1. Romero R, Mazor M, Sepulveda W, Avila C, Copeland D, and Williams J (1992). Tumor necrosis factor in preterm and term labor. Am. J. Obstet. Gynecol 166, 1576–1587.
    1. Romero R, Dey SK, and Fisher SJ (2014a). Preterm labor: one syndrome, many causes. Science 345, 760–765.
    1. Romero R, Miranda J, Chaiworapongsa T, Chaemsaithong P, Gotsch F, Dong Z, Ahmed AI, Yoon BH, Hassan SS, Kim CJ, et al. (2014b). A novel molecular microbiologic technique for the rapid diagnosis of microbial invasion of the amniotic cavity and intra-amniotic infection in preterm labor with intact membranes. Am. J. Reprod. Immunol 71, 330–358.
    1. Romero R, Miranda J, Chaiworapongsa T, Korzeniewski SJ, Chaemsaithong P, Gotsch F, Dong Z, Ahmed AI, Yoon BH, Hassan SS, et al. (2014c). Prevalence and clinical significance of sterile intra-amniotic inflammation in patients with preterm labor and intact membranes. Am. J. Reprod. Immunol 72, 458–474.
    1. Romero R, Grivel JC, Tarca AL, Chaemsaithong P, Xu Z, Fitzgerald W, Hassan SS, Chaiworapongsa T, and Margolis L (2015). Evidence of perturbations of the cytokine network in preterm labor. Am. J. Obstet. Gynecol 213, 836.e1–836.e18.
    1. Romero R, Chaemsaithong P, Chaiyasit N, Docheva N, Dong Z, Kim CJ, Kim YM, Kim JS, Qureshi F, Jacques SM, et al. (2017). CXCL10 and IL-6: Markers of two different forms of intra-amniotic inflammation in preterm labor. Am. J. Reprod. Immunol 78, e12685.
    1. Rowe JH, Ertelt JM, Aguilera MN, Farrar MA, and Way SS (2011). Foxp3(+) regulatory T cell expansion required for sustaining pregnancy compromises host defense against prenatal bacterial pathogens. Cell Host Microbe 10, 54–64.
    1. Rowe JH, Ertelt JM, Xin L, and Way SS (2012). Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature 490, 102–106.
    1. Saito S, Nakashima A, Shima T, and Ito M (2010). Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am. J. Reprod. Immunol 63, 601–610.
    1. Salafia CM, Vogel CA, Vintzileos AM, Bantham KF, Pezzullo J, and Silberman L (1991). Placental pathologic findings in preterm birth. Am. J. Obstet. Gynecol 165, 934–938.
    1. Salvany-Celades M, van der Zwan A, Benner M, Setrajcic-Dragos V, Bougleux Gomes HA, Iyer V, Norwitz ER, Strominger JL, and Tilburgs T (2019). Three Types of Functional Regulatory T Cells Control T Cell Responses at the Human Maternal-Fetal Interface. Cell Rep. 27, 2537–2547.
    1. Samstein RM, Josefowicz SZ, Arvey A, Treuting PM, and Rudensky AY (2012). Extrathymic generation of regulatory T cells in placental mammals mitigates maternal-fetal conflict. Cell 150, 29–38.
    1. Sasaki Y, Sakai M, Miyazaki S, Higuma S, Shiozaki A, and Saito S (2004). Decidual and peripheral blood CD4+CD25+ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol. Hum. Reprod 10, 347–353.
    1. Schober L, Radnai D, Schmitt E, Mahnke K, Sohn C, and Steinborn A (2012). Term and preterm labor: decreased suppressive activity and changes in composition of the regulatory T-cell pool. Immunol. Cell Biol 90, 935–944.
    1. Shima T, Sasaki Y, Itoh M, Nakashima A, Ishii N, Sugamura K, and Saito S (2010). Regulatory T cells are necessary for implantation and maintenance of early pregnancy but not late pregnancy in allogeneic mice. J. Reprod. Immunol 85, 121–129.
    1. Shima T, Inada K, Nakashima A, Ushijima A, Ito M, Yoshino O, and Saito S (2015). Paternal antigen-specific proliferating regulatory T cells are increased in uterine-draining lymph nodes just before implantation and in pregnant uterus just after implantation by seminal plasma-priming in allogeneic mouse pregnancy. J. Reprod. Immunol 108, 72–82.
    1. Shiozaki A, Yoneda S, Yoneda N, Yonezawa R, Matsubayashi T, Seo G, and Saito S (2014). Intestinal microbiota is different in women with preterm birth: results from terminal restriction fragment length polymorphism analysis. PLoS ONE 9, e111374.
    1. Sindram-Trujillo A, Scherjon S, Kanhai H, Roelen D, and Claas F (2003). Increased T-cell activation in decidua parietalis compared to decidua basalis in uncomplicated human term pregnancy. Am. J. Reprod. Immunol 49, 261–268.
    1. Sindram-Trujillo AP, Scherjon SA, van Hulst-van Miert PP, Kanhai HH, Roelen DL, and Claas FH (2004). Comparison of decidual leukocytes following spontaneous vaginal delivery and elective cesarean section in uncomplicated human term pregnancy. J. Reprod. Immunol 62, 125–137.
    1. Slutsky R, Romero R, Xu Y, Galaz J, Miller D, Done B, Tarca AL, Gregor S, Hassan SS, Leng Y, and Gomez-Lopez N (2019). Exhausted and Senescent T Cells at the Maternal-Fetal Interface in Preterm and Term Labor. J. Immunol. Res 2019, 3128010.
    1. Soneson C, Love MI, and Robinson MD (2015). Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res. 4, 1521.
    1. Sorokin Y, Romero R, Mele L, Wapner RJ, Iams JD, Dudley DJ, Spong CY, Peaceman AM, Leveno KJ, Harper M, et al. (2010). Maternal serum interleukin-6, C-reactive protein, and matrix metalloproteinase-9 concentrations as risk factors for preterm birth <32 weeks and adverse neonatal outcomes. Am. J. Perinatol 27, 631–640.
    1. Sotiriadis A, Hernandez-Andrade E, da Silva Costa F, Ghi T, Glanc P, Khalil A, Martins WP, Odibo AO, Papageorghiou AT, Salomon LJ, and Thilaganathan B; ISUOG CSC Pre-eclampsia Task Force (2019). ISUOG Practice Guidelines: role of ultrasound in screening for and follow-up of pre-eclampsia. Ultrasound Obstet. Gynecol 53, 7–22.
    1. St Louis D, Romero R, Plazyo O, Arenas-Hernandez M, Panaitescu B, Xu Y, Milovic T, Xu Z, Bhatti G, Mi QS, et al. (2016). Invariant NKT Cell Activation Induces Late Preterm Birth That Is Attenuated by Rosiglitazone. J. Immunol 196, 1044–1059.
    1. Steinborn A, Engst M, Haensch GM, Mahnke K, Schmitt E, Meuer S, and Sohn C (2010). Small for gestational age (SGA) neonates show reduced suppressive activity of their regulatory T cells. Clin. Immunol 134, 188–197.
    1. Steinborn A, Schmitt E, Kisielewicz A, Rechenberg S, Seissler N, Mahnke K, Schaier M, Zeier M, and Sohn C (2012). Pregnancy-associated diseases are characterized by the composition of the systemic regulatory T cell (Treg) pool with distinct subsets of Tregs. Clin. Exp. Immunol 167, 84–98.
    1. Svensson J, Jenmalm MC, Matussek A, Geffers R, Berg G, and Ernerudh J (2011). Macrophages at the fetal-maternal interface express markers of alternative activation and are induced by M-CSF and IL-10. J. Immunol 187, 3671–3682.
    1. Svensson-Arvelund J, Mehta RB, Lindau R, Mirrasekhian E, Rodriguez-Martinez H, Berg G, Lash GE, Jenmalm MC, and Ernerudh J (2015). The human fetal placenta promotes tolerance against the semiallogeneic fetus by inducing regulatory T cells and homeostatic M2 macrophages. J. Immunol 194, 1534–1544.
    1. Tilburgs T, Roelen DL, van der Mast BJ, van Schip JJ, Kleijburg C, de Groot-Swings GM, Kanhai HHH, Claas FHJ, and Scherjon SA (2006). Differential distribution of CD4(+)CD25(bright) and CD8(+)CD28(−) T-cells in decidua and maternal blood during human pregnancy. Placenta 27 (Suppl A), S47–S53.
    1. Tilburgs T, Roelen DL, van der Mast BJ, de Groot-Swings GM, Kleijburg C, Scherjon SA, and Claas FH (2008). 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 180, 5737–5745.
    1. Tsuda S, Zhang X, Hamana H, Shima T, Ushijima A, Tsuda K, Muraguchi A, Kishi H, and Saito S (2018). Clonally Expanded Decidual Effector Regulatory T Cells Increase in Late Gestation of Normal Pregnancy, but Not in Preeclampsia, in Humans. Front. Immunol 9, 1934.
    1. Tsuda S, Nakashima A, Shima T, and Saito S (2019). New Paradigm in the Role of Regulatory T Cells During Pregnancy. Front. Immunol 10, 573.
    1. Vacca P, Montaldo E, Croxatto D, Loiacono F, Canegallo F, Venturini PL, Moretta L, and Mingari MC (2015). Identification of diverse innate lymphoid cells in human decidua. Mucosal Immunol. 8, 254–264.
    1. van der Zwan A, Bi K, Norwitz ER, Crespo AC, Claas FHJ, Strominger JL, and Tilburgs T (2018). Mixed signature of activation and dysfunction allows human decidual CD8+ T cells to provide both tolerance and immunity. Proc. Natl. Acad. Sci. USA 115, 385–390.
    1. Wei X, Zhang J, Gu Q, Huang M, Zhang W, Guo J, and Zhou X (2017). Reciprocal Expression of IL-35 and IL-10 Defines Two Distinct Effector Treg Subsets that Are Required for Maintenance of Immune Tolerance. Cell Rep. 21, 1853–1869.
    1. Willcockson AR, Nandu T, Liu CL, Nallasamy S, Kraus WL, and Mahendroo M (2018). Transcriptome signature identifies distinct cervical pathways induced in lipopolysaccharide-mediated preterm birth. Biol. Reprod 98, 408–421.
    1. Xiong H, Zhou C, and Qi G (2010). Proportional changes of CD4+CD25+Foxp3+ regulatory T cells in maternal peripheral blood during pregnancy and labor at term and preterm. Clin. Invest. Med 33, E422.
    1. Xu Y, Plazyo O, Romero R, Hassan SS, and Gomez-Lopez N (2015). Isolation of Leukocytes from the Human Maternal-fetal Interface. J. Vis. Exp 99, e52863.
    1. Xu Y, Romero R, Miller D, Kadam L, Mial TN, Plazyo O, Garcia-Flores V, Hassan SS, Xu Z, Tarca AL, et al. (2016). An M1-like Macrophage Polarization in Decidual Tissue during Spontaneous Preterm Labor That Is Attenuated by Rosiglitazone Treatment. J. Immunol 196, 2476–2491.
    1. Xu Y, Romero R, Miller D, Silva P, Panaitescu B, Theis KR, Arif A, Hassan SS, and Gomez-Lopez N (2018). Innate lymphoid cells at the human maternal-fetal interface in spontaneous preterm labor. Am. J. Reprod. Immunol 79, e12820.
    1. Yao Z, Painter SL, Fanslow WC, Ulrich D, Macduff BM, Spriggs MK, and Armitage RJ (1995). Human IL-17: a novel cytokine derived from T cells. J. Immunol 155, 5483–5486.
    1. Yellon SM (2017). Contributions to the dynamics of cervix remodeling prior to term and preterm birth. Biol. Reprod 96, 13–23.
    1. Yoon BH, Romero R, Moon JB, Shim SS, Kim M, Kim G, and Jun JK (2001). Clinical significance of intra-amniotic inflammation in patients with preterm labor and intact membranes. Am. J. Obstet. Gynecol 185, 1130–1136.
    1. Zenclussen AC, Gerlof K, Zenclussen ML, Sollwedel A, Bertoja AZ, Ritter T, Kotsch K, Leber J, and Volk HD (2005). Abnormal T-cell reactivity against paternal antigens in spontaneous abortion: adoptive transfer of pregnancy-induced CD4+CD25+ T regulatory cells prevents fetal rejection in a murine abortion model. Am. J. Pathol 166, 811–822.
    1. Zhao K, Zhao D, Huang D, Yin L, Chen C, Pan B, Wu Q, Li Z, Yao Y, Shen E, et al. (2014). Interleukin-22 aggravates murine acute graft-versus-host disease by expanding effector T cell and reducing regulatory T cell. J. Interferon Cytokine Res 34, 707–715.

Source: PubMed

3
Sottoscrivi