DOI QR코드

DOI QR Code

Epithelial to mesenchymal transition (EMT) of feto-maternal reproductive tissues generates inflammation: a detrimental factor for preterm birth

  • Menon, Ramkumar (Division of Basic and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston)
  • Received : 2021.12.05
  • Accepted : 2022.05.12
  • Published : 2022.08.31

Abstract

Human pregnancy is a delicate and complex process where multiorgan interactions between two independent systems, the mother, and her fetus, maintain pregnancy. Intercellular interactions that can define homeostasis at the various cellular level between the two systems allow uninterrupted fetal growth and development until delivery. Interactions are needed for tissue remodeling during pregnancy at both fetal and maternal tissue layers. One of the mechanisms that help tissue remodeling is via cellular transitions where epithelial cells undergo a cyclic transition from epithelial to mesenchymal (EMT) and back from mesenchymal to epithelial (MET). Two major pregnancy-associated tissue systems that use EMT, and MET are the fetal membrane (amniochorion) amnion epithelial layer and cervical epithelial cells and will be reviewed here. EMT is often associated with localized inflammation, and it is a well-balanced process to facilitate tissue remodeling. Cyclic transition processes are important because a terminal state or the static state of EMT can cause accumulation of proinflammatory mesenchymal cells in the matrix regions of these tissues and increase localized inflammation that can cause tissue damage. Interactions that determine homeostasis are often controlled by both endocrine and paracrine mediators. Pregnancy maintenance hormone progesterone and its receptors are critical for maintaining the balance between EMT and MET. Increased intrauterine oxidative stress at term can force a static (terminal) EMT and increase inflammation that are physiologic processes that destabilize homeostasis that maintain pregnancy to promote labor and delivery of the fetus. However, conditions that can produce an untimely increase in EMT and inflammation can be pathologic. These tissue damages are often associated with adverse pregnancy complications such as preterm prelabor rupture of the membranes (pPROM) and spontaneous preterm birth (PTB). Therefore, an understanding of the biomolecular processes that maintain cyclic EMT-MET is critical to reducing the risk of pPROM and PTB. Extracellular vesicles (exosomes of 40-160 nm) that can carry various cargo are involved in cellular transitions as paracrine mediators. Exosomes can carry a variety of biomolecules as cargo. Studies specifically using exosomes from cells undergone EMT can carry a pro-inflammatory cargo and in a paracrine fashion can modify the neighboring tissue environment to cause enhancement of uterine inflammation.

Keywords

Acknowledgement

This review is supported by 1R01HD100729 (NIH/NICHD) to Dr. Ramkumar Menon.

References

  1. Romero R, Dey SK and Fisher SJ (2014) Preterm labor: one syndrome, many causes. Science 345, 760-765 https://doi.org/10.1126/science.1251816
  2. Tal R and Taylor HS (2000) Endocrinology of Pregnancy; in Endotext, Feingold KR, Anawalt B, Boyce A et al. (eds.), South Dartmouth (MA)
  3. Rokas A, Mesiano S, Tamam O, LaBella A, Zhang G and Muglia L (2020) Developing a theoretical evolutionary framework to solve the mystery of parturition initiation. Elife 9, e58343
  4. Smith R (1998) Alterations in the hypothalamic pituitary adrenal axis during pregnancy and the placental clock that determines the length of parturition. J Reprod Immunol 39, 215-220 https://doi.org/10.1016/S0165-0378(98)00023-0
  5. Menon R, Bonney EA, Condon J, Mesiano S and Taylor RN (2016) Novel concepts on pregnancy clocks and alarms: redundancy and synergy in human parturition. Hum Reprod Update 22, 535-560 https://doi.org/10.1093/humupd/dmw022
  6. Mendelson CR, Montalbano AP and Gao L (2017) Fetal-to-maternal signaling in the timing of birth. J Steroid Biochem Mol Biol 170, 19-27 https://doi.org/10.1016/j.jsbmb.2016.09.006
  7. Beck S, Wojdyla D, Say L et al (2010) The worldwide incidence of preterm birth: a systematic review of maternal mortality and morbidity. Bull World Health Organ 88, 31-38 https://doi.org/10.2471/BLT.08.062554
  8. Shapiro-Mendoza CK and Lackritz EM (2012) Epidemiology of late and moderate preterm birth. Semin Fetal Neonatal Med 17, 120-125 https://doi.org/10.1016/j.siny.2012.01.007
  9. Simmons LE, Rubens CE, Darmstadt GL and Gravett MG (2010) Preventing preterm birth and neonatal mortality: exploring the epidemiology, causes, and interventions. Semin Perinatol 34, 408-415 https://doi.org/10.1053/j.semperi.2010.09.005
  10. Blencowe H, Cousens S, Oestergaard MZ et al (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 https://doi.org/10.1016/S0140-6736(12)60820-4
  11. Dammann O and Leviton A (1997) Maternal intrauterine infection, cytokines, and brain damage in the preterm newborn. Pediatr Res 42, 1-8 https://doi.org/10.1203/00006450-199707000-00001
  12. Jacobsson B (2004) Infectious and inflammatory mechanisms in preterm birth and cerebral palsy. Eur J Obstet Gynecol Reprod Biol 115, 159-160 https://doi.org/10.1016/j.ejogrb.2003.11.014
  13. Muglia LJ and Katz M (2010) The enigma of spontaneous preterm birth. N Engl J Med 362, 529-535 https://doi.org/10.1056/NEJMra0904308
  14. McCabe ER, Carrino GE, Russell RB and Howse JL (2014) Fighting for the next generation: US Prematurity in 2030. Pediatrics 134, 1193-1199 https://doi.org/10.1542/peds.2014-2541
  15. Villar J, Papageorghiou AT, Knight HE et al (2012) The preterm birth syndrome: a prototype phenotypic classification. Am J Obstet Gynecol 206, 119-123 https://doi.org/10.1016/j.ajog.2011.10.866
  16. Parry S and Strauss JF, 3rd (1998) Premature rupture of the fetal membranes. N Engl J Med 338, 663-670 https://doi.org/10.1056/NEJM199803053381006
  17. Shubert PJ, Diss E and Iams JD (1992) Etiology of preterm premature rupture of membranes. Obstet Gynecol Clin North Am 19, 251-263 https://doi.org/10.1016/S0889-8545(21)00348-X
  18. Naeye RL and Peters EC (1980) Causes and consequences of premature rupture of fetal membranes. Lancet 1, 192-194 https://doi.org/10.1016/S0140-6736(80)90674-1
  19. French JI and McGregor JA (1996) The pathobiology of premature rupture of membranes. Semin Perinatol 20, 344-368 https://doi.org/10.1016/S0146-0005(96)80002-4
  20. Menon R and Fortunato SJ (2007) Infection and the role of inflammation in preterm premature rupture of the membranes. Best Pract Res Clin Obstet Gynaecol 21, 467-478 https://doi.org/10.1016/j.bpobgyn.2007.01.008
  21. Elovitz MA and Mrinalini C (2004) Animal models of preterm birth. Trends Endocrinol Metab 15, 479-487 https://doi.org/10.1016/j.tem.2004.10.009
  22. Chauhan SP and Ananth CV (2013) Periviable births: epidemiology and obstetrical antecedents. Semin Perinatol 37, 382-388 https://doi.org/10.1053/j.semperi.2013.06.020
  23. Lawn JE, Kinney MV, Belizan JM et al (2013) Born too soon: accelerating actions for prevention and care of 15 million newborns born too soon. Reprod Health 10 Suppl 1, S6
  24. Pavlicev M and Norwitz ER (2018) Human parturition: nothing more than a delayed menstruation. Reprod Sci 25, 166-173 https://doi.org/10.1177/1933719117725830
  25. Hay ED (1981) Extracellular matrix. J Cell Biol 91, 205s-223s https://doi.org/10.1083/jcb.91.3.205s
  26. Bourne GL and LACY D (1960) Ultra-structure of human amnion and its possible relation to the circulation of amniotic fluid. Nature 186, 952-954 https://doi.org/10.1038/186952a0
  27. Castillo-Castrejon M, Meraz-Cruz N, Gomez-Lopez N et al (2014) Choriodecidual cells from term human pregnancies show distinctive functional properties related to the induction of labor 2. Am J Reprod Immunol 71, 86-93 https://doi.org/10.1111/aji.12179
  28. King AE, Kelly RW, Sallenave JM, Bocking AD and Challis JR (2007) Innate immune defences in the human uterus during pregnancy 6. Placenta 28, 1099-1106 https://doi.org/10.1016/j.placenta.2007.06.002
  29. Vora S, Abbas A, Kim CJ et al (2010) Nuclear factor-kappa B localization and function within intrauterine tissues from term and preterm labor and cultured fetal membranes 2. Reprod Biol Endocrinol 8, 8
  30. Montenegro D, Romero R, Kim SS et al (2009) Expression patterns of microRNAs in the chorioamniotic membranes: a role for microRNAs in human pregnancy and parturition 3. J Pathol 217, 113-121 https://doi.org/10.1002/path.2463
  31. Menon R, Richardson LS and Lappas M (2019) Fetal membrane architecture, aging and inflammation in pregnancy and parturition. Placenta 79, 40-45 https://doi.org/10.1016/j.placenta.2018.11.003
  32. Richardson LS, Vargas G, Brown T et al (2017) Discovery and characterization of human amniochorionic membrane microfractures. Am J Pathol 187, 2821-2830 https://doi.org/10.1016/j.ajpath.2017.08.019
  33. Richardson LS, Taylor RN and Menon R (2020) Reversible EMT and MET mediate amnion remodeling during pregnancy and labor. Sci Signal 13, 1486
  34. Richardson LS, Radnaa E, Urrabaz-Garza R, Lavu N and Menon R (2020) Stretch, scratch, and stress: suppressors and supporters of senescence in human fetal membranes. Placenta 99, 27-34 https://doi.org/10.1016/j.placenta.2020.07.013
  35. Jacobs SO, Sheller-Miller S, Richardson LS, Urrabaz-Garza R, Radnaa E and Menon R (2020) Characterizing the immune cell population in the human fetal membrane. Am J Reprod Immunol 85, e13368
  36. Vadillo-Ortega F, Gonzalez-Avila G, Furth EE et al (1995) 92-kd type IV collagenase (matrix metalloproteinase-9) activity in human amniochorion increases with labor. Am J Pathol 146, 148-156
  37. Vadillo-Ortega F, Hernandez A, Gonzalez-Avila G, Bermejo L, Iwata K and Strauss JF, III (1996) Increased matrix metalloproteinase activity and reduced tissue inhibitor of metalloproteinases-1 levels in amniotic fluids from pregnancies complicated by premature rupture of membranes. Am J Obstet Gynecol 174, 1371-1376 https://doi.org/10.1016/S0002-9378(96)70687-7
  38. Vadillo-Ortega F, Sadowsky DW, Haluska GJ et al (2002) Identification of matrix metalloproteinase-9 in amniotic fluid and amniochorion in spontaneous labor and after experimental intrauterine infection or interleukin-1 beta infusion in pregnant rhesus monkeys. Am J Obstet Gynecol 186, 128-138 https://doi.org/10.1067/mob.2002.118916
  39. Bryant-Greenwood GD (1998) The extracellular matrix of the human fetal membranes: structure and function. Placenta 19, 1-11 https://doi.org/10.1016/S0143-4004(98)90092-3
  40. Strauss JF 3rd (2013) Extracellular matrix dynamics and fetal membrane rupture. Reprod Sci 20, 140-153 https://doi.org/10.1177/1933719111424454
  41. Mahendroo M (2012) Cervical remodeling in term and preterm birth: insights from an animal model. Reproduction 143, 429-438 https://doi.org/10.1530/REP-11-0466
  42. Yellon SM (2017) Contributions to the dynamics of cervix remodeling prior to term and preterm birth. Biol Reprod 96, 13-23 https://doi.org/10.1095/biolreprod.116.142844
  43. Read CP, Word RA, Ruscheinsky MA, Timmons BC and Mahendroo MS (2007) Cervical remodeling during pregnancy and parturition: molecular characterization of the softening phase in mice. Reproduction 134, 327-340 https://doi.org/10.1530/REP-07-0032
  44. Word RA, Li XH, Hnat M and Carrick K (2007) Dynamics of cervical remodeling during pregnancy and parturition: mechanisms and current concepts. Semin Reprod Med 25, 69-79 https://doi.org/10.1055/s-2006-956777
  45. Becher N, Adams Waldorf K, Hein M and Uldbjerg N (2009) The cervical mucus plug: structured review of the literature. Acta Obstet Gynecol Scand 88, 502-513 https://doi.org/10.1080/00016340902852898
  46. Critchfield AS, Yao G, Jaishankar A et al (2013) Cervical mucus properties stratify risk for preterm birth. PLoS One 8, e69528
  47. Hansen LK, Becher N, Bastholm S et al (2014) The cervical mucus plug inhibits, but does not block, the passage of ascending bacteria from the vagina during pregnancy. Acta Obstet Gynecol Scand 93, 102-108 https://doi.org/10.1111/aogs.12296
  48. Frew L, Makieva S, McKinlay AT et al (2014) Human cathelicidin production by the cervix. PLoS One 9, e103434
  49. Yellon SM (2019) Immunobiology of cervix ripening. Front Immunol 10, 3156
  50. Sennstrom MB, Ekman G, Westergren-Thorsson G et al (2000) Human cervical ripening, an inflammatory process mediated by cytokines. Mol Hum Reprod 6, 375-381 https://doi.org/10.1093/molehr/6.4.375
  51. Stygar D, Wang H, Vladic YS, Ekman G, Eriksson H and Sahlin L (2002) Increased level of matrix metalloproteinases 2 and 9 in the ripening process of the human cervix. Biol Reprod 67, 889-894 https://doi.org/10.1095/biolreprod.102.005116
  52. Iwahashi M, Muragaki Y, Ooshima A and Umesaki N (2003) Decreased type I collagen expression in human uterine cervix during pregnancy. J Clin Endocrinol Metab 88, 2231-2235 https://doi.org/10.1210/jc.2002-021213
  53. Myers DA (2012) The recruitment and activation of leukocytes into the immune cervix: further support that cervical remodeling involves an immune and inflammatory mechanism. Biol Reprod 87, 107
  54. Zucker S, Hymowitz M, Conner C et al (1999) Measurement of matrix metalloproteinases and tissue inhibitors of metalloproteinases in blood and tissues. Clinical and experimental applications. Ann N Y Acad Sci 878, 212-227 https://doi.org/10.1111/j.1749-6632.1999.tb07687.x
  55. Matrisian LM, Wright J, Newell K and Witty JP (1994) Matrix-degrading metalloproteinases in tumor progression. Princess Takamatsu Symp 24, 152-161
  56. Tantengco OAG, Richardson LS, Vink J et al (2021) Progesterone alters human cervical epithelial and stromal cell transition and migration: Implications in cervical remodeling during pregnancy and parturition. Mol Cell Endocrinol 529, 111276
  57. Chai M, Barker G, Menon R and Lappas M (2012) Increased oxidative stress in human fetal membranes overlying the cervix from term non-labouring and post labour deliveries. Placenta 33, 604-610 https://doi.org/10.1016/j.placenta.2012.04.014
  58. Gonzalez JM, Franzke CW, Yang F, Romero R and Girardi G (2011) Complement activation triggers metalloproteinases release inducing cervical remodeling and preterm birth in mice. Am J Pathol 179, 838-849 https://doi.org/10.1016/j.ajpath.2011.04.024
  59. Gonzalez JM, Dong Z, Romero R and Girardi G (2011) Cervical remodeling/ripening at term and preterm delivery: the same mechanism initiated by different mediators and different effector cells. PLoS One 6, e26877
  60. Moore RM, Mansour JM, Redline RW, Mercer BM and Moore JJ (2006) The physiology of fetal membrane rupture: insight gained from the determination of physical properties. Placenta 27, 1037-1051 https://doi.org/10.1016/j.placenta.2006.01.002
  61. Becher N, Hein M, Danielsen CC and Uldbjerg N (2004) Matrix metalloproteinases and their inhibitors in the cervical mucus plug at term of pregnancy. Am J Obstet Gynecol 191, 1232-1239 https://doi.org/10.1016/j.ajog.2004.03.023
  62. Vadillo-Ortega F and Estrada-Gutierrez G (2005) Role of matrix metalloproteinases in preterm labour. BJOG 112 Suppl 1, 19-22 https://doi.org/10.1111/j.1471-0528.2005.00579.x
  63. Wang H, Parry S, Macones G et al (2004) Functionally significant SNP MMP8 promoter haplotypes and preterm premature rupture of membranes (PPROM). Hum Mol Genet 13, 2659-2669 https://doi.org/10.1093/hmg/ddh287
  64. Jeong HC, Kim HY, Kim HY et al (2021) Changes in gene expression of cervical collagens, metalloproteinases, and tissue inhibitors of metalloproteinases after partial cervical excision-induced preterm labor in mice. PLoS One 16, e0250108
  65. Watari M, Watari H, DiSanto ME, Chacko S, Shi GP and Strauss JF 3rd (1999) Pro-inflammatory cytokines induce expression of matrix-metabolizing enzymes in human cervical smooth muscle cells. Am J Pathol 154, 1755-1762 https://doi.org/10.1016/S0002-9440(10)65431-4
  66. Rath W, Winkler M and Kemp B (1998) The importance of extracellular matrix in the induction of preterm delivery. J Perinat Med 26, 437-441 https://doi.org/10.1515/jpme.1998.26.6.437
  67. Akins ML, Luby-Phelps K, Bank RA and Mahendroo M (2011) Cervical softening during pregnancy: regulated changes in collagen cross-linking and composition of matricellular proteins in the mouse. Biol Reprod 84, 1053-1062 https://doi.org/10.1095/biolreprod.110.089599
  68. Menon R and Fortunato SJ (2004) The role of matrix degrading enzymes and apoptosis in rupture of membranes. J Soc Gynecol Investig 11, 427-437 https://doi.org/10.1016/j.jsgi.2004.04.001
  69. Fortunato SJ and Menon R (2002) Screening of novel matrix metalloproteinases (MMPs) in human fetal membranes. J Assist Reprod Genet 19, 483-486 https://doi.org/10.1023/A:1020362519981
  70. Maymon E, Romero R, Pacora P et al (2001) A role for the 72 kDa gelatinase (MMP-2) and its inhibitor (TIMP-2) in human parturition, premature rupture of membranes and intraamniotic infection. J Perinat Med 29, 308-316
  71. Fortunato SJ, Menon R and Lombardi SJ (1999) MMP/TIMP imbalance in amniotic fluid during PROM: an indirect support for endogenous pathway to membrane rupture. J Perinat Med 27, 362-368
  72. Richardson L and Menon R (2018) Proliferative, migratory, and transition properties reveal metastate of human amnion cells. Am J Pathol 188, 2004-2015 https://doi.org/10.1016/j.ajpath.2018.05.019
  73. Menon R and Richardson LS (2017) Preterm prelabor rupture of the membranes: a disease of the fetal membranes. Semin Perinatol 41, 409-419 https://doi.org/10.1053/j.semperi.2017.07.012
  74. Kalluri R and Weinberg RA (2009) The basics of epithelial-mesenchymal transition. J Clin Invest 119, 1420-1428 https://doi.org/10.1172/JCI39104
  75. Kalluri R (2009) EMT: when epithelial cells decide to become mesenchymal-like cells. J Clin Invest 119, 1417-1419 https://doi.org/10.1172/JCI39675
  76. Janzen C, Sen S, Lei MY, Gagliardi de Assumpcao M, Challis J and Chaudhuri G (2017) The role of epithelial to mesenchymal transition in human amniotic membrane rupture. J Clin Endocrinol Metab 102, 1261-1269
  77. Mogami H, Hari Kishore A, Akgul Y and Word RA (2017) Healing of preterm ruptured fetal membranes. Sci Rep 7, 13139
  78. Richardson L, Dixon CL, Aguilera-Aguirre L and Menon R (2018) Oxidative stress-induced TGF-beta/TAB1-mediated p38MAPK activation in human amnion epithelial cells. Biol Reprod 99, 1100-1112 https://doi.org/10.1093/biolre/ioy135
  79. Menon R, Boldogh I, Urrabaz-Garza R et al (2013) Senescence of primary amniotic cells via oxidative DNA damage. PLoS One 8, e83416
  80. Lavu N, Richardson L, Radnaa E et al (2019) Oxidative stress-induced downregulation of glycogen synthase kinase 3 beta in fetal membranes promotes cellular senescencedagger. Biol Reprod 101, 1018-1030 https://doi.org/10.1093/biolre/ioz119
  81. Omere C, Richardson L, Saade GR, Bonney EA, Kechichian T and Menon R (2020) Interleukin (IL)-6: a friend or foe of pregnancy and parturition? evidence from functional studies in fetal membrane cells. Front Physiol 11, 891
  82. Lozovyy V, Richardson L, Saade G and Menon R (2021) Progesterone receptor membrane components: key regulators of fetal membrane integrity. Biol Reprod 104, 445-456 https://doi.org/10.1093/biolre/ioaa192
  83. Menon R and Moore JJ (2020) Fetal membranes, not a mere appendage of the placenta, but a critical part of the fetal-maternal interface controlling parturition. Obstet Gynecol Clin North Am 47, 147-162 https://doi.org/10.1016/j.ogc.2019.10.004
  84. Murtha AP and Menon R (2015) Regulation of fetal membrane inflammation: a critical step in reducing adverse pregnancy outcome. Am J Obstet Gynecol 213, 447-448 https://doi.org/10.1016/j.ajog.2015.07.008
  85. Dutta EH, Behnia F, Boldogh I et al (2016) Oxidative stress damage-associated molecular signaling pathways differentiate spontaneous preterm birth and preterm premature rupture of the membranes. Mol Hum Reprod 22, 143-157 https://doi.org/10.1093/molehr/gav074
  86. Polettini J, Dutta EH, Behnia F, Saade GR, Torloni MR and Menon R (2015) Aging of intrauterine tissues in spontaneous preterm birth and preterm premature rupture of the membranes: a systematic review of the literature. Placenta 36, 969-973 https://doi.org/10.1016/j.placenta.2015.05.003
  87. Menon R, Behnia F, Polettini J and Richardson LS (2020) Novel pathways of inflammation in human fetal membranes associated with preterm birth and preterm prelabor rupture of the membranes. Semin Immunopathol 42, 431-450 https://doi.org/10.1007/s00281-020-00808-x
  88. Tantengco OAG, Richardson LS and Menon R (2021) Effects of a gestational level of estradiol on cellular transition, migration, and inflammation in cervical epithelial and stromal cells. Am J Reprod Immunol 85, e13370
  89. Tantengco OAG and Menon R (2020) Contractile function of the cervix plays a role in normal and pathological pregnancy and parturition. Med Hypotheses 145, 110336
  90. Tantengco OAG, Vink J, Medina PMB and Menon R (2021) Oxidative stress promotes cellular damages in the cervix: implications for normal and pathologic cervical function in human pregnancydagger. Biol Reprod 105, 204-216 https://doi.org/10.1093/biolre/ioab058
  91. Mackler AM, Iezza G, Akin MR, McMillan P and Yellon SM (1999) Macrophage trafficking in the uterus and cervix precedes parturition in the mouse. Biol Reprod 61, 879-883 https://doi.org/10.1095/biolreprod61.4.879
  92. Polettini J, Dutta EH, Behnia F, Saade GR, Torloni MR and Menon R (2015) Aging of intrauterine tissues in spontaneous preterm birth and preterm premature rupture of the membranes: a systematic review of the literature. Placenta 36, 969-973 https://doi.org/10.1016/j.placenta.2015.05.003
  93. Martin LF, Moco NP, de Lima MD et al (2017) Histologic chorioamnionitis does not modulate the oxidative stress and antioxidant status in pregnancies complicated by spontaneous preterm delivery. BMC Pregnancy Childbirth 17, 376
  94. Burton GJ and Jauniaux E (2004) Placental oxidative stress: from miscarriage to preeclampsia. J Soc Gynecol Investig 11, 342-352 https://doi.org/10.1016/j.jsgi.2004.03.003
  95. Myatt L (2010) Review: reactive oxygen and nitrogen species and functional adaptation of the placenta. Placenta 31 Suppl, S66-S69 https://doi.org/10.1016/j.placenta.2009.12.021
  96. Woods JR Jr (2001) Reactive oxygen species and preterm premature rupture of membranes-a review. Placenta 22 Suppl A, S38-S44 https://doi.org/10.1053/plac.2001.0638
  97. Dennery PA (2010) Oxidative stress in development: nature or nurture? Free Radic Biol Med 49, 1147-1151 https://doi.org/10.1016/j.freeradbiomed.2010.07.011
  98. Sahlin L, Wang H, Stjernholm Y et al (2000) The expression of glutaredoxin is increased in the human cervix in term pregnancy and immediately post-partum, particularly after prostaglandin-induced delivery. Mol Hum Reprod 6, 1147-1153 https://doi.org/10.1093/molehr/6.12.1147
  99. Ryu HK, Moon JH, Heo HJ, Kim JW and Kim YH (2017) Maternal c-reactive protein and oxidative stress markers as predictors of delivery latency in patients experiencing preterm premature rupture of membranes. Int J Gynaecol Obstet 136, 145-150 https://doi.org/10.1002/ijgo.12024
  100. Heng YJ, Di Quinzio MK, Permezel M, Rice GE and Georgiou HM (2010) Temporal expression of antioxidants in human cervicovaginal fluid associated with spontaneous labor. Antioxid Redox Signal 13, 951-957 https://doi.org/10.1089/ars.2010.3122
  101. Barrios De Tomasi J, Opata MM and Mowa CN (2019) Immunity in the cervix: interphase between immune and cervical epithelial cells. J Immunol Res 2019, 7693183
  102. Yarbrough VL, Winkle S and Herbst-Kralovetz MM (2015) Antimicrobial peptides in the female reproductive tract: a critical component of the mucosal immune barrier with physiological and clinical implications. Hum Reprod Update 21, 353-377 https://doi.org/10.1093/humupd/dmu065
  103. da Fonseca EB, Damiao R and Nicholaides K (2009) Prevention of preterm birth based on short cervix: progesterone. Semin Perinatol 33, 334-337 https://doi.org/10.1053/j.semperi.2009.06.006
  104. Romero R, Nicolaides K, Conde-Agudelo A et al (2012) Vaginal progesterone in women with an asymptomatic sonographic short cervix in the midtrimester decreases preterm delivery and neonatal morbidity: a systematic review and metaanalysis of individual patient data. Am J Obstet Gynecol 206, 124-119
  105. Romero R, Yeo L, Chaemsaithong P, Chaiworapongsa T and Hassan SS (2014) Progesterone to prevent spontaneous preterm birth 5. Semin. Fetal Neonatal Med 19, 15-26 https://doi.org/10.1016/j.siny.2013.10.004
  106. Berghella V and Saccone G (2019) Cervical assessment by ultrasound for preventing preterm delivery. Cochrane Database Syst Rev 9, CD007235
  107. Yost NP and Cox SM (2000) Infection and preterm labor. Clin Obstet Gynecol 43, 759-767 https://doi.org/10.1097/00003081-200012000-00006
  108. Holst RM, Jacobsson B, Hagberg H and Wennerholm UB (2006) Cervical length in women in preterm labor with intact membranes: relationship to intra-amniotic inflammation/microbial invasion, cervical inflammation and preterm delivery. Ultrasound Obstet Gynecol 28, 768-774 https://doi.org/10.1002/uog.3837
  109. Holst RM, Mattsby-Baltzer I, Wennerholm UB, Hagberg H and Jacobsson B (2005) Interleukin-6 and interleukin-8 in cervical fluid in a population of Swedish women in preterm labor: relationship to microbial invasion of the amniotic fluid, intra-amniotic inflammation, and preterm delivery. Acta Obstet Gynecol Scand 84, 551-557 https://doi.org/10.1111/j.0001-6349.2005.00708.x
  110. Aaltone R, Jalava J, Laurikainen E, Karkkainen U and Alanen A (2002) Cervical ureaplasma urealyticum colonization: comparison of PCR and culture for its detection and association with preterm birth. Scand J Infect Dis 34, 35-40 https://doi.org/10.1080/00365540110077074
  111. Menon R and Shahin H (2020) Extracellular vesicles in spontaneous preterm birth. Am J Reprod Immunol 85, e13353
  112. Shepherd MC, Radnaa E, Tantengco OA et al (2021) Extracellular vesicles from maternal uterine cells exposed to risk factors cause fetal inflammatory response. Cell Commun Signal 19, 100
  113. Hadley EE, Sheller-Miller S, Saade G et al (2018) Amnion epithelial cell derived exosomes induce inflammatory changes in uterine cells. Am J Obstet Gynecol 219, 478
  114. Monsivais LA, Sheller Miller S, Russell W et al (2020) Fetal membrane extracellular vesicle profiling reveals distinct pathways induced by infection and inflammation in vitro. Am J Reprod Immunol 84, e13282
  115. Tantengco OAG, Radnaa E, Shahin H, Kechichian T and Menon R (2021) Cross talk: trafficking and functional impact of maternal exosomes at the Feto-maternal Interface under normal and pathologic states. Biol Reprod 105, 1562-1576 https://doi.org/10.1093/biolre/ioab181
  116. Shahin HI, Radnaa E, Tantengco OAG et al (2021) Microvesicles and exosomes released by amnion epithelial cells under oxidative stress cause inflammatory changes in uterine cellsdagger. Biol Reprod 105, 464-480 https://doi.org/10.1093/biolre/ioab088
  117. Sheller-Miller S, Choi K, Choi C and Menon R (2019) Cyclic-recombinase-reporter mouse model to determine exosome communication and function during pregnancy. Am J Obstet Gynecol 221, 502 e501-502 e512
  118. Sheller-Miller S, Lei J, Saade G, Salomon C, Burd I and Menon R (2016) Feto-maternal trafficking of exosomes in murine pregnancy models. Front Pharmacol 7, 432
  119. Sheller-Miller S, Trivedi J, Yellon SM and Menon R (2019) Exosomes cause preterm birth in mice: evidence for paracrine signaling in pregnancy. Sci Rep 9, 608
  120. Menon R, Mesiano S and Taylor RN (2017) Programmed fetal membrane senescence and exosome-mediated signaling: a mechanism associated with timing of human parturition. Front Endocrinol (Lausanne) 8, 196