The role of fetal membranes during gestation, at term, and preterm labor | Placenta and Reproductive Medicine

The role of fetal membranes during gestation, at term, and preterm labor

Fetal membranes role during pregnancy

Authors

  • Nina Truong The John Sealy School of Medicine, University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA
  • Lauren Richardson Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA
  • Ramkumar Menon Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA

DOI:

https://doi.org/10.54844/prm.2022.0296

Keywords:

amnion, chorion, membrane integrity, preterm rupture of the membrane, labor signaling

Abstract

During pregnancy, the fetal membranes (i.e., amniochorionic membranes) surround the intrauterine cavity and provide mechanical, immune, and endocrine support to protect the fetus. Though they are a vital component of the intrauterine cavity, the fetal membranes are largely overlooked as an extension of the placenta, leading to a poor understanding of their role during gestation, parturition, or preterm birth. The fetal membranes are comprised of fetal cellular and stromal layers and line up with maternal decidua forming the feto-maternal interface during pregnancy. This interface plays a large role during pregnancy and the induction of term or preterm parturition (e.g., labor). Here we summarize the function of the fetal membranes focusing on their role during gestation at term, and during preterm births.

Author Biographies

Nina Truong, The John Sealy School of Medicine, University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA

 

 

 

Lauren Richardson, Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA

 

 

 

Ramkumar Menon, Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA

 

 

 

References

Mauri A, Perrini M, Mateos JM, et al. Second harmonic generation microscopy of fetal membranes under deformation: normal and altered morphology. Placenta. 2013;34(11):1020–1026. DOI: https://doi.org/10.1016/j.placenta.2013.09.002

Mauri A, Ehret AE, Perrini M, et al. Deformation mechanisms of human amnion: quantitative studies based on second harmonic generation microscopy. J Biomech. 2015;48(9):1606–1613. DOI: https://doi.org/10.1016/j.jbiomech.2015.01.045

Mauri A, Perrini M, Ehret AE, De Focatiis DSA, Mazza E. Timedependent mechanical behavior of human amnion: macroscopic and microscopic characterization. Acta Biomater. 2015;11:314–323. DOI: https://doi.org/10.1016/j.actbio.2014.09.012

Perrini M, Mauri A, Ehret AE, et al. Mechanical and microstructural investigation of the cyclic behavior of human amnion. J Biomech Eng. 2015;137(6):061010. DOI: https://doi.org/10.1115/1.4030054

Kumar D, Moore RM, Mercer BM, Mansour JM, Redline RW, Moore JJ. The physiology of fetal membrane weakening and rupture: insights gained from the determination of physical properties revisited. Placenta. 2016;42:59–73. DOI: https://doi.org/10.1016/j.placenta.2016.03.015

Moore RM, Mansour JM, Redline RW, Mercer BM, Moore JJ. The physiology of fetal membrane rupture: insight gained from the determination of physical properties. Placenta. 2006;27(11–12):1037–1051. DOI: https://doi.org/10.1016/j.placenta.2006.01.002

Richardson L, Vargas G, Brown T, et al. Redefining 3Dimensional placental membrane microarchitecture using multiphoton microscopy and optical clearing. Placenta. 2017;53:66–75.

Menon R, Lappas M, Zakar T. Editorial: the role of the fetal membranes in pregnancy and birth. Front Physiol. 2021;12:653084. DOI: https://doi.org/10.3389/fphys.2021.653084

Zhu C, Jiang Z, Johnson GA, Burghardt RC, Bazer FW, Wu G. Nutritional and physiological regulation of water transport in the conceptus. Adv Exp Med Biol. 2022;1354:109–125. DOI: https://doi.org/10.1007/978-3-030-85686-1_6

Seo MJ, Lim JH, Kim DH, Bae HR. Loss of aquaporin-3 in placenta and fetal membranes induces growth restriction in mice. Dev Reprod. 2018;22(3):263–273. DOI: https://doi.org/10.12717/DR.2018.22.3.263

Ducza E, Csányi A, Gáspár R. Aquaporins during pregnancy: their function and significance. Int J Mol Sci. 2017;18(12):2593. DOI: https://doi.org/10.3390/ijms18122593

Kammala A, Benson M, Ganguly E, et al. Fetal membranes contribute to drug transport across the feto-maternal interface utilizing the breast cancer resistance protein (BCRP). Life (Basel). 2022;12(2):166. DOI: https://doi.org/10.3390/life12020166

Ganguly E, Kammala AK, Benson M, Richardson LS, Han A, Menon R. Organic anion transporting polypeptide 2B1 in human fetal membranes: a novel gatekeeper for drug transport during pregnancy? Front Pharmacol. 2021;12:771818. DOI: https://doi.org/10.3389/fphar.2021.771818

Kumar D, Springel E, Moore RM, et al. Progesterone inhibits in vitro fetal membrane weakening. Am J Obstet Gynecol. 2015;213(4):520.e1–520. e9. DOI: https://doi.org/10.1016/j.ajog.2015.06.014

Merlino A, Welsh T, Erdonmez T, et al. Nuclear progesterone receptor expression in the human fetal membranes and decidua at term before and after labor. Reprod Sci. 2009;16(4):357–363. DOI: https://doi.org/10.1177/1933719108328616

Lozovyy V, Richardson L, Saade G, Menon R. Progesterone receptor membrane components: key regulators of fetal membrane integrity. Biol Reprod. 2021;104(2):445–456. DOI: https://doi.org/10.1093/biolre/ioaa192

Richardson LS, Taylor RN, Menon R. Reversible EMT and MET mediate amnion remodeling during pregnancy and labor. Sci Signal. 2020;13(618):eaay1486. DOI: https://doi.org/10.1126/scisignal.aay1486

Fortunato SJ, Menon R, Lombardi SJ, LaFleur B. Interleukin-10 inhibition of gelatinases in fetal membranes: therapeutic implications in preterm premature rupture of membranes. Obstet Gynecol. 2001;98(2):284–288. DOI: https://doi.org/10.1097/00006250-200108000-00018

Hadley EE, Sheller-Miller S, Saade G, et al. Amnion epithelial cellderived exosomes induce inflammatory changes in uterine cells. Am J Obstet Gynecol. 2018;219(5):478.e1–478478.e21. DOI: https://doi.org/10.1016/j.ajog.2018.08.021

Duchesne MJ, Thaler-Dao H, de Paulet AC. Prostaglandin synthesis in human placenta and fetal membranes. Prostaglandins. 1978;15(1):19–42. DOI: https://doi.org/10.1016/S0090-6980(78)80003-3

Keirse MJ, Turnbull AC. The fetal membranes as a possible source of amniotic fluid prostaglandins. Br J Obstet Gynaecol. 1976;83(2):146–151. DOI: https://doi.org/10.1111/j.1471-0528.1976.tb00797.x

Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986–1000. DOI: https://doi.org/10.1161/ATVBAHA.110.207449

Brown NL, Alvi SA, Elder MG, Bennett PR, Sullivan MH. The regulation of prostaglandin output from term intact fetal membranes by anti-inflammatory cytokines. Immunology. 2000;99(1):124–133. DOI: https://doi.org/10.1046/j.1365-2567.2000.00942.x

Gibb W. The role of prostaglandins in human parturition. Ann Med. 1998;30(3):235–241. DOI: https://doi.org/10.3109/07853899809005850

Mistry HD, Williams PJ. The importance of antioxidant micronutrients in pregnancy. Oxid Med Cell Longev. 2011;2011:841749. DOI: https://doi.org/10.1155/2011/841749

Menon R, Richardson LS. Preterm prelabor rupture of the membranes: a disease of the fetal membranes. Semin Perinatol. 2017;41(7):409–419. DOI: https://doi.org/10.1053/j.semperi.2017.07.012

Menon R, Behnia F, Polettini J, Saade GR, Campisi J, Velarde M. Placental membrane aging and HMGB1 signaling associated with human parturition. Aging. 2016;8(2):216–230. DOI: https://doi.org/10.18632/aging.100891

Sheller S, Papaconstantinou J, Urrabaz-Garza R, et al. Amnion-epithelialcell- derived exosomes demonstrate physiologic state of cell under oxidative stress. PLoS One. 2016;11(6):e0157614. DOI: https://doi.org/10.1371/journal.pone.0157614

Menon R. Menon R. Human fetal membranes at term: Dead tissue or signalers of parturition? Placenta. 2016;44:1–5. Vora S, Abbas A, Kim CJ, et al. DOI: https://doi.org/10.1016/j.placenta.2016.05.013

Nuclear factor-kappa B localization and function within intrauterine tissues from term and preterm labor and cultured fetal membranes. Reprod Biol Endocrinol. 2010;8:8.

Menon R, Boldogh I, Urrabaz-Garza R, et al. Senescence of primary amniotic cells via oxidative DNA damage. PLoS One. 2013;8(12):e83416.

Choltus H, Lavergne M, Belville C, et al. Occurrence of a RAGEmediated inflammatory response in human fetal membranes. Front Physiol. 2020;11:581. DOI: https://doi.org/10.3389/fphys.2020.00581

Lappas M. NOD1 and NOD2 regulate proinflammatory and prolabor mediators in human fetal membranes and myometrium via nuclear factor-kappa B. Biol Reprod. 2013;89(1):14. DOI: https://doi.org/10.1095/biolreprod.113.110056

Lappas M, Riley C, Lim R, et al. MAPK and AP-1 proteins are increased in term pre-labour fetal membranes overlying the cervix: regulation of enzymes involved in the degradation of fetal membranes. Placenta. 2011;32(12):1016–1025. DOI: https://doi.org/10.1016/j.placenta.2011.09.011

Vora S, Abbas A, Kim CJ, et al. Nuclear factor-kappa B localization and function within intrauterine tissues from term and preterm labor and cultured fetal membranes. Reprod Biol Endocrinol. 2010;8:8. DOI: https://doi.org/10.1186/1477-7827-8-8

Martin L, Richardson L, Menon R. Characteristics, properties, and functionality of fetal membranes: an overlooked area in the field of parturition. In: Skinner MK, ed. Encyclopedia of Reproduction. 2nd ed. Elsevier; 2018: 387–398. DOI: https://doi.org/10.1016/B978-0-12-801238-3.64498-7

Richardson LS, Vargas G, Brown T, et al. Discovery and characterization of human amniochorionic membrane microfractures. Am J Pathol. 2017;187(12):2821–2830. DOI: https://doi.org/10.1016/j.ajpath.2017.08.019

Sheller-Miller S, Urrabaz-Garza R, Saade G, Menon R. Damage-Associated molecular pattern markers HMGB1 and cell-Free fetal telomere fragments in oxidative-Stressed amnion epithelial cell-Derived exosomes. J Reprod Immunol. 2017;123:3–11. DOI: https://doi.org/10.1016/j.jri.2017.08.003

Mercer BM. Preterm premature rupture of the membranes. Obstet Gynecol. 2003;101(1):178–193. DOI: https://doi.org/10.1016/S0029-7844(02)02366-9

Joyce EM, Moore JJ, Sacks MS. Biomechanics of the fetal membrane prior to mechanical failure: review and implications. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S121–S127. DOI: https://doi.org/10.1016/j.ejogrb.2009.02.014

Dayal S, Hong PL. Premature Rupture Of Membranes. In: StatPearls. StatPearls Publishing; 2022.

Koh CT, Tonsomboon K, Oyen ML. Fracture toughness of human amniotic membranes. Interface Focus. 2019;9(5):20190012. DOI: https://doi.org/10.1098/rsfs.2019.0012

Dars S, Malik S, Samreen I, Kazi RA. Maternal morbidity and perinatal outcome in preterm premature rupture of membranes before 37 weeks gestation. Pak J Med Sci. 2014;30(3):626–629. DOI: https://doi.org/10.12669/pjms.303.4853

Medina TM, Hill DA. Preterm premature rupture of membranes: diagnosis and management. Am Fam Physician. 2006;73(4):659–664.

Dutta EH, Behnia F, Boldogh I, et al. Oxidative stress damage-associated molecular signaling pathways differentiate spontaneous preterm birth and preterm premature rupture of the membranes. Mol Hum Reprod. 2016;22(2):143–157. DOI: https://doi.org/10.1093/molehr/gav074

Menon R, Boldogh I, Urrabaz-Garza R, et al. Senescence of primary amniotic cells via oxidative DNA damage. PLoS One. 2013;8(12):e83416. DOI: https://doi.org/10.1371/journal.pone.0083416

Menon R. Oxidative stress damage as a detrimental factor in preterm birth pathology. Front Immunol. 2014;5:567. DOI: https://doi.org/10.3389/fimmu.2014.00567

Bredeson S, Papaconstantinou J, Deford JH, et al. HMGB1 promotes a p38MAPK associated non-infectious inflammatory response pathway in human fetal membranes. PLoS One. 2014;9(12):e113799. DOI: https://doi.org/10.1371/journal.pone.0113799

Menon R, Boldogh I, Hawkins HK, et al. Histological evidence of oxidative stress and premature senescence in preterm premature rupture of the human fetal membranes recapitulated

in vitro. Am J Pathol. 2014;184(6):1740–1751. DOI: https://doi.org/10.1016/j.ajpath.2014.02.011

Richardson L, Vargas G, Brown T, et al. Redefining 3Dimensional placental membrane microarchitecture using multiphoton microscopy and optical clearing. Placenta. 2017;53:66–75. DOI: https://doi.org/10.1016/j.placenta.2017.03.017

Gibbs RS, Duff P. Progress in pathogenesis and management of clinical intraamniotic infection. Am J Obstet Gynecol. 1991;164(5):1317–1326. DOI: https://doi.org/10.1016/0002-9378(91)90707-X

Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol. 1993;36(4):795–808. DOI: https://doi.org/10.1097/00003081-199312000-00004

Yoon BH, Romero R, Moon JB, et al. Clinical significance of intraamniotic inflammation in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2001;185(5):1130–1136. DOI: https://doi.org/10.1067/mob.2001.117680

Masucci MT, Minopoli M, Del Vecchio S, Carriero MV. The emerging role of neutrophil extracellular traps (NETs) in tumor progression and metastasis. Front Immunol. 2020;11:1749. DOI: https://doi.org/10.3389/fimmu.2020.01749

Goulopoulou S, Davidge ST. Molecular mechanisms of maternal vascular dysfunction in preeclampsia. Trends Mol Med. 2015;21(2):88–97. DOI: https://doi.org/10.1016/j.molmed.2014.11.009

Craige SM, Kant S, Keaney JF Jr. Reactive oxygen species in endothelial function - from disease to adaptation-. Circ J. 2015;79(6):1145–1155. DOI: https://doi.org/10.1253/circj.CJ-15-0464

Veiga ECA, Korkes HA, Salomão KB, Cavalli RC. Association of LEPTIN and other inflammatory markers with preeclampsia: a systematic review. Front Pharmacol. 2022;13:966400. DOI: https://doi.org/10.3389/fphar.2022.966400

Roberts JM, Lain KY. Recent insights into the pathogenesis of preeclampsia. Placenta. 2002;23(5):359–372. DOI: https://doi.org/10.1053/plac.2002.0819

Litwiniuk M, Grzela T. Amniotic membrane: new concepts for an old dressing. Wound Repair Regen. 2014;22(4):451–456. DOI: https://doi.org/10.1111/wrr.12188

Fuchs F, Riis P. Antenatal sex determination. Nature. 1956;177(4503):330. DOI: https://doi.org/10.1038/177330a0

Simpson JL, Elias S. Isolating fetal cells in maternal circulation for prenatal diagnosis. Prenat Diagn. 1994;14(13):1229–1242. DOI: https://doi.org/10.1002/pd.1970141308

Published

2023-03-20

How to Cite

1.
Truong N, Richardson L, Menon R. The role of fetal membranes during gestation, at term, and preterm labor: Fetal membranes role during pregnancy. PRM. 2023;2. doi:10.54844/prm.2022.0296

Issue

Section

Review Article

Categories

Downloads

Download data is not yet available.
REVIEW ARTICLE

The role of fetal membranes during gestation, at term, and preterm labor


Nina Truong1, Ramkumar Menon2, Lauren Richardson2

1The John Sealy School of Medicine, University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA;

2Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA


Corresponding Author:

Dr. Lauren Richardson, E-mail: lestaffo@utmb.edu; https://orcid.org/0000-0001-8392-2833


Received: 20 December 2022 Accepted: 28 February 2023 Published: 20 March 2023


ABSTRACT

During pregnancy, the fetal membranes (i.e., amniochorionic membranes) surround the intrauterine cavity and provide mechanical, immune, and endocrine support to protect the fetus. Though they are a vital component of the intrauterine cavity, the fetal membranes are largely overlooked as an extension of the placenta, leading to a poor understanding of their role during gestation, parturition, or preterm birth. The fetal membranes are comprised of fetal cellular and stromal layers and line up with maternal decidua forming the feto-maternal interface during pregnancy. This interface plays a large role during pregnancy and the induction of term or preterm parturition (e.g., labor). Here we summarize the function of the fetal membranes focusing on their role during gestation at term, and during preterm births.

Key words: amnion, chorion, membrane integrity, preterm rupture of the membrane, labor signaling

THE FUNCTION OF THE FETAL MEMBRANE DURING GESTATION

The fetal membrane is a thick, collagenous bilayer comprised of a fetal amnion and chorion epithelium connected to the maternal decidua capsularis. Acting as the feto-maternal interface, one of the fetal membranes’ primary functions during gestation is to compartmentalize potential maternal exposures to various risk factors (e.g., maternal infection, environmental pollutants, behavioral risks) that may induce a fetal inflammatory response that can lead to adverse pregnancy outcomes if not contained. The amnion component of the fetal membrane predominantly contributes to this barrier function by: (1) supporting fetal growth and amniotic fluid load, (2) withstanding local deformation and mechanical stretch,[1-6] (3) preserving an intact amnion epithelial layer,[7] (4) testing the environment and responding to local insults (e.g., infection, reactive oxygen species), and (5) undergoing cellular and collagen remodeling to maintain extracellular matrix homeostasis. All of these roles are critical for maintaining membrane integrity during pregnancy (Figure 1). Conversely, the multi-layered chorion is responsible for modulating the choriodecidual (feto-maternal) interface immune environment by producing anti-inflammatory hormones and cytokines that buffer maternal immune cell invasion. Chorion expression of human leukocyte antigens (HLA-G) provides immune tolerance at this interface. These endocrine (a major source of progesterone) and paracrine signalers (anti-inflammatory interleukin-10) help to maintain immune cell homeostasis at the choriodecidual interface during gestation (Figure 1).

Figure 1

Figure 1. Feto-maternal interfaces’ functions during gestation. The fetal membrane lines the intrauterine cavity and creates a protective barrier around the fetus throughout gestation. It is divided into two components: the amnion membrane (amnion epithelial cells [blue]– stroma [green lines]– amnion mesenchymal cells [light purple]) and the chorion membrane (chorion mesenchymal cells [dark purple] – stroma [green lines]– chorion trophoblast [yellow]), that are connected to the maternal decidua to create the feto-maternal interface. During gestation, it is critical that the mechanical, immune, and inflammatory properties of the membranes stay in homeostasis to allow for normal gestation progression. Both the amnion and chorion membranes play a role in these functions by contributing to cellular transitions (epithelial-to-mesenchymal and mesenchymal-to-epithelial transitions [black arrows]) to replace shed cells in both layers and to produce collagen (dark green lines). Additionally, the chorion regulates the quiescence of resident decidua immune cells (orange) by expressing HLA receptors (dark blue) (i.e., HLA-G) and by secreting progesterone. To maintain inflammatory homeostasis, all cell layers in both membranes regulate the amount of pro-inflammatory cytokines and oxidative stress within the intrauterine cavity by producing anti-inflammatory cytokines and breaking down free oxygen radicles. HLA: human leukocyte antigen.

Additionally, the fetal membrane acts as a critical site of communication between the mother and the fetus.[8,9] This is evident by the roles it plays in maintaining immune homeostasis and transporting nutrients, water, and drugs during gestation.[1013] Inflammatory balance and the regulation of immune homeostasis at the fetal membrane decidual interface are essential components in preventing membrane dysfunction. Progesterone is a key anti-inflammatory gestational hormone that mediates inflammatory homeostasis by acting through an inducible negative feedback system that exists locally at the feto-maternal interface.[9,14] During gestation, the placenta and fetal membrane cells produce progesterone resulting in endocrine signaling through its nuclear (i.e., progesterone receptor isoform A [PRA] and progesterone receptor isoform B [PRB][15]) and membrane-bound progesterone receptors (i.e., PGRMC1/2 and MPα/MPβ[16]) across both feto-maternal interfaces. To note, the amnion and chorion layers do not contain nuclear progesterone receptors; however, they do express PGRMC1 and 2. Progesterone signaling during gestation is critical for the amnion epithelium to maintain its barrier function of sequestering the amniotic fluid in the intrauterine cavity. It does this by promoting mesenchymal cells within the fetal membrane collagen layers to undergo cellular transitions (i.e., mesenchymal-to-epithelial transition [MET[17]]) (Figure 1) to refill weak points in the epithelium. Progesterone also plays a critical role in inhibiting the function of pro-inflammatory cytokines (i.e., granulocyte-macrophage colony-stimulating factor [GM-CSF], tumor necrosis factor α [TNF-α], Interleukin [IL]-6, IL-8) that can weaken collagen in the fetal membranes[6,14]. Anti-inflammatory cytokine IL-10, produced locally by fetal membrane cells, has also been shown to be functionally similar to progesterone in its ability to regulate pro-inflammatory cytokine production and HLA-G expression contributing to the maintenance of immune homeostasis.[8,18]

Water and nutrient transport during gestation are essential for maintaining normal growth and development of the fetus[9,11] Aquaporins are plasma membrane proteins existing at more than eleven different isoforms whose levels in the endometrium and fetal-derived cells are altered during the implantation process.[11] Aquaporins have been shown to play a role in myometrial contractions and cervical ripening; However, they do contribute to the following process at the feto-maternal interface: (1) transport of water across the placenta to support the placentation, (2) maintenance of amniotic fluid volume, (3) and embryonic/fetal survival, growth, and development.[9-11] It has also been proposed that fetal membrane cells might express amnio acids, fatty acids, and vitamin transport proteins, similar to the placenta, suggesting fetal membranes could also play a role in nutrient transport to the fetus during gestation.

Recent studies have also identified the presence of transport proteins (involved primarily in the transport of various molecules, including drugs from the mother) within fetal membrane cells and in fetal membrane cell-derived vesicles (i.e., exosomes—extracellular vesicles of 30–160 nm released from these cells).[13,19] A few of these transporter proteins include permeability-glycoprotein (P-gp), breast cancer resistance protein-1 (BCRP-1), and the organic anion transporting polypeptide 2B1 (OATP2B1) were primarily localized in the chorion with limited expression in the amnion.[12,13] Their expression and function in the fetal membranes are similar to that observed in the placenta, changing the dogma that the placenta is the only intrauterine organ responsible for molecule transport.[12] Further studies and a better understanding of this drug transportation route may lead to novel therapeutic approaches and innovative drug therapies during pregnancy. This is critical, as a breakdown of the homeostatic processes of the fetal membranes described above leads to catastrophic membrane failure. This failure and dysfunction leading to fetal membrane rupture is a physiologic requirement at term but pathologic and consequential if it happens prematurely.

FETAL MEMBRANES’ CONTRIBUTION TO LABOR INDUCTION

While the placenta is critical for fetal survival during gestation and at term, it is reported to not play a prominent role in the induction of labor signaling as unhindered placental function is required until fetal delivery. Conversely, the fetal membranes induce multiple parallel signaling pathways described below to shift immune homeostasis to a pro-inflammatory environment that contributes to labor onset. Discovered in 1976 by Keirse et al., and verified in 1978 by Duchesne et al., it was initially thought that the membranes' sole contribution to labor onset was prostaglandin production.[20,21] Prostaglandins are a group of lipid autacoids derived from arachidonic acid synthesized as a response to local tissue damage or infection that has been well known to control inflammation, vascular flow, and the induction of labor.[22] Labor-inducing prostaglandins produced by the fetal membranes and decidua, such as prostaglandin E2 (PGE2) and prostaglandin F (PGF), are vital mediators of parturition that act primarily through collagen degradation to alter membrane structure, contract the myometrium, and ripen the cervix.[20,23] These lipids are found at elevated amounts in the amniotic fluid at both term and preterm parturition.[20,2224] These elevated levels may result from decreased prostaglandin metabolism, increased prostaglandin production, and or changes in the expression of different prostaglandin receptors.[24] The multifactorial regulation of prostaglandins is a crucial component of labor signaling. However, it is only one of the many pathways fetal membrane cells induce to ensure the delivery of the fetus. Various pathways will be discussed in greater detail in the following paragraphs.

Close to term, the growing fetus ensues increased aerobic respiration and metabolic demands. These changes, accompanied by low levels of fetal and maternal antioxidants (i.e., selenium, copper, zinc, manganese, vitamin C, and vitamin E[25]), yield elevated amounts of reactive oxygen species (ROS) in the amniotic cavity (Figure 2). The imbalance between ROS production and levels of antioxidants leads to intraamniotic oxidative stress (OS) buildup. Increased ROS induces cellular, organelle, and DNA damage within fetal membrane cells, causing them to undergo telomere alteration. Telomere alteration activates stress signaler p38 mitogen-activated protein kinase activation (MAPK) that promotes cellular senescence (i.e., aging). While the rapid and abundant indication of fetal membrane senescence may also be induced pathologically, this process is usually physiologic and coincides with fetal maturation at term. In parallel, intrauterine OS and cytokines (i.e., transforming growth factor [TGF] β) accelerate fetal membrane weakening by inducing cellular transition dysfunction (i.e., increased epithelial-to-mesenchymal transition [EMT] and inhibition of MET through PGRMC functional withdrawal)[17] (Figure 2) in the amnion cells that leads to an increase in pro-inflammatory mesenchymal cells, collagen breakdown, and a lower mechanical threshold for rupture.[26] Senescent fetal membrane cells also exhibit senescence-associated secretory phenotype (SASP), a unique group of inflammatory mediators comprising cytokines, chemokines, and various pro-inflammatory markers. SASPs precede the release of pro-inflammatory damage-associated molecular patterns (DAMPs) such as high mobility group box 1 and cell-free fetal telomere fragments.[2729] SASP and DAMPS promote labor by propagating senescence across the feto-maternal interface and by increasing the inflammatory load (Figure 2). Changes in inflammatory homeostasis trigger the uterine tissues to transition toward a pro-labor phenotype. These pathways have been demonstrated within term labor[29,30] and preterm birth29,30 fetal membranes, as well as in vitro cell models treated with OS inducers.[31] Additionally, there are other parallel signaling cascades (i.e., toll-like receptors, the receptor for advanced glycation end products,[32] nucleotide oligomerization domain,[33] AP-1,[34] and NF-κB,[35]) that play roles in promoting a pro-inflammatory environment within the intrauterine cavity at term.

Figure 2

Figure 2. Labor-associated changes at the feto-maternal interface. Cartoon schematic showing fetal membrane anatomy (amnion epithelial cells [blue] – stroma [green lines] – amnion mesenchymal cells [light purple]) and the chorion membrane (chorion mesenchymal cells [dark purple] – stroma [green lines] – chorion trophoblast [yellow] – maternal decidua [green] and processes that contribute to parturition signaling. At term, oxidative stress (yellow stars) contributes to the induction of amnion membrane weakening (i.e., EMT [red lined cells], collagen break down, senescence) and chorion barrier degradation (i.e., HLA downregulation [dark blue receptors], immune cell infiltration, and decreased hormone production). Both membrane components increase the production of inflammation and propagate inflammatory signals from the fetal to maternal tissue. These signals can be propagated through migrating cells or exosomes (grey dots) that carry cellular cargo to neighboring cells/tissues that then induce oxidative stress. HLA: human leukocyte antigen; EMT: epithelial-to-mesenchymal transition; SASP: senescence-associated secretory phenotype; DAMP: damage-associated molecular patterns.

Propagation of these signals from fetal to the maternal compartments could occur in two different ways: diffusion through (1) microfractures (i.e., fissures in fetal membrane collagen layers connecting the feto-maternal compartments), or (2) exosomes released from fetal membrane cells (Figure 2). Microfractures have been shown to be in higher quantity and contain deeper features within term or preterm fetal membranes.[26,36,37] These findings have been recreated experimentally in vitro, suggesting their relevance in the propagation of parturition signals.[37] In parallel, fetal membrane cell-derived exosomes are capable of carrying contents from the cell of their origin to distant cell layers.[19,28,38] It is reported that exosome cargo from OS fetal membrane cells contains active forms of p38MAPK and SASPs capable of promoting labor signaling. Thus, fetal membranes at term undergo senescence and cellular transitions, weaken the collagen layers, and produce powerful pro-inflammatory signals sent through various channels to convey to the maternal side the readiness for labor and delivery of the fetus (Figure 2).

FETAL MEMBRANE PATHOLOGIES CORRELATED WITH PRETERM BIRTH

Similar labor induction pathways can be activated physiologically at term or pathologically preterm. Preterm birth (PTB) is defined as birth before the 37th week of complete gestation. Preterm labor and delivery can be medically induced or spontaneous. Spontaneous preterm birth can be subdivided into two categories: preceded by preterm labor with intact membranes or preceded by preterm premature rupture of membranes (pPROM). Among spontaneous PTBs, pPROM or failure of the fetal membrane is the initiating cause of 40% of preterm births and affects 3% of all total pregnancies.[39,40] pPROM is classified as a rupture of the membranes before the onset of labor and before 37 weeks of gestation[1,2,39,41,42] (Figure 3). Fetal membrane rupture, at any stage of gestation, can be induced by both physical stretch or oxidative stress that biochemically weakens the collagen layers of the membranes leading to catastrophic mechanical failure. Rupture of the membranes increases the risk of ascending intraamniotic infection and adverse fetal outcomes such as hypoxia, pulmonary hypoplasia, respiratory distress syndrome, neonatal sepsis, umbilical cord prolapse, necrotizing enterocolitis, neurosensory impairment, cerebral palsy, developmental delay, and even fetal death.[39,43,44] The etiologies, pathways, and mechanisms of spontaneous PTB and pPROM are still unclear. However, it has been shown that spontaneous PTB is associated with minimal DNA damage, extracellular signal-regulated kinases and c-Jun N-terminal kinases activation, which does not lead to premature cellular senescecne.[45] While in cases of pPROM, OS and inflammation (i.e., ROS, thrombin, and pro-inflammatory cytokines)[14,46,47] arising from fetal membrane signaling cascades (i.e., Activator protein 1, NF-κB, p38MAPK),[34,45,48] induce senescence,[49] cellular transitions (i.e., EMT and microfractures),[17,37,50] and paracrine or endocrine exosomal signaling tht can play critical roles in premature labor onset. Further investigation is needed to delineate the synergistic and differential roles of each step in the labor cascade between physiological term parturition and pathologic preterm labor.

Figure 3

Figure 3. Schematic of the feto-maternal interface during adverse pregnancy outcomes. The intrauterine cavity (pink) is surrounded by maternal (decidua [green]) and fetal (amnion [blue] and chorion [yellow]) layers that form the feto-maternal interface during pregnancy. During adverse pregnancy complication that results in ascending vaginal infection (i.e., bacteria/viruses [red dots]), infectious agents transverse the decidua-fetal membrane interface inducing chorioamnionitis. This results in an infiltration of immune cells, specifically neutrophils, that migrate through the feto-maternal interface and induce large amounts of inflammation. As the infection progresses, infectious agents that reach the amnionic cavity create what is called an intraamniotic infection which is correlated with preterm birth. Infection-induced preterm birth is also associated with pPROM. This occurs due to infectious or oxidative stress-induced cell disruption and collagen degradation leading to weakening and rupture of the membranes. pPROM: preterm premature rupture of the membranes.

Histologic chorioamnionitis (HCA) is a relatively common pregnancy complication that can lead to adverse maternal and infant outcomes. Clinically HCA presents as maternal fever, uterine fundal tenderness, maternal tachycardia (> 100 beats/min), fetal tachycardia (> 160 beats/min), and purulent or malodorous amniotic fluid.[51,52] Additionally, it has become increasingly linked to preterm delivery, with 40%–70% of HCA births complicated by pPROM or spontaneous PTB.[53] HCA is often associated with intraamniotic polymicrobial bacterial infection (i.e., Group B streptococcus, Mycoplasma pneumoniae, Ureaplasma, Gardnerella vaginalis, Escherichia coli, and Bacteroides) and or increased sterile inflammation (i.e., ROS) within the intrauterine cavity. HCA presents as a gradient promotion of migrating immune cells, dominantly neutrophils, from the maternal circulation or decidua capsularis stimulated into the fetal membranes by chemokines (i.e., IL-8 and GM-CSF). These chemokines, along with bacteria antigens, activate neutrophils which produce pro-inflammatory cytokines that activate prostaglandins and collagen-degrading enzymes (i.e., MMPs) as they migrate from the chorion to the amnion. In parallel, within the collagen of the fetal membranes, neutrophils release neutrophil extracellular traps (i.e., NETs), which are web-like structures formed of DNA fibers, histones, and antimicrobial proteins that trap pathogens. These NETs immobilize the bacterial threat but adversely induce NETosis, a very specific type of NET-induced cell death that can promote collagen degradation through laminin cleavage and remodeling.[54] Pro-inflammatory cytokine production and NET formation lead to fetal membrane weakening and subsequent rupture preterm. The high prevalence of HCA, especially compared to the rarer incidence of placentitis (i.e., immune cell infiltration of the placenta), makes this condition, and hence the fetal membrane, an essential subject of study for PTB onset.

Gestational diseases, such as pre-eclampsia and gestational diabetes, play a role in many adverse maternal outcomes by inducing systemic stress (i.e., high ROS, downregulation of NADPH oxidases)[55,56] and inflammation (i.e., Interluken-6, Tumor necrosis factor, Leptin).[57] Pre-eclampsia is a leading cause of maternal and fetal morbidity and mortality defined by stage 2 hypertension with onset after 20 weeks of gestation in a woman with previously normal blood pressure. Gestational diabetes, characterized by a diagnosis of diabetes mellitus for the first time during pregnancy, has been identified as a risk factor for pre-eclampsia. Though the pathophysiology of pre-eclampsia has not been fully understood, it has been suggested that systemic maternal endothelial dysfunction resulting from abnormal placental development, preexisting maternal risk factors (i.e., pre-pregnancy obesity, advanced maternal age, insulin resistance, chronic hypertension, etc.),[58] and the physiological activation of the inflammatory cascade caused by normal pregnancy may lead to OS, inflammation, and vascular dysfunction. Though these diseases typically affect the placenta, and hence are maternally targeted, systemic inflammation induced by pre-eclampsia could negatively affect fetal membrane function leading to preterm rupture. Assessing the biological function of fetal membranes and or fetal membrane cellular layers will provide a more thorough understanding of the mechanisms by which the fetal membranes functions in normal pregnancy environment and its potential contributions to the onset preterm birth. This knowledge is expected to provide novel strategies for treating or diagnosing adverse pregnancy outcomes.

DIAGNOSTIC POTENTIAL OF FETAL MEMBRANE COMPONENTS

Since the early 19th century, fetal membrane tissue and its cellular components have been utilized in a variety of clinical fields to treat ocular surface defects and heal diabetic ulcers, surgical incisions, burns, deep wounds, and leg ulcers.[59] However, it was not until 1956 that fetal membrane components were used as diagnostics, when Fuchs and Riis et al. isolated fetal trophoblast cells (e.g., fetal membrane and placenta in origin) from amniocytes to identify fetal sex and genetic disorters.[60] This invasive technique has now been predominantly replaced with isolations of fetal trophoblasts from the maternal blood in the 1990’s to diagnose chromosomal abnormalities (i.e., trisomy 21, trisomy 18, and Klinefelter syndrome), fetal sex, mendelian disorders, and fetal rhesus blood type.[61] The future of prenatal diagnosis lies in merging the information known about labor-associated biological processes with a non-invasive sampling of maternal blood, leading to fetal membrane component isolation. Such tools would allow clinicians to sample fetal membrane cells and or exosomes from maternal blood to (1) detect biomarkers of adverse pregnancy outcomes, (2) assess fetal membrane cellular and inflammatory status, and (3) identify fetal genetic and epigenetic changes due to maternal environmental exposures.

SUMMARY

The fetal membranes play a vital role in the labor cascade by undergoing tissue and cellular-specific changes that lead to the propagation of labor-initiating signals from the fetus to the mother at term and preterm. As the feto-maternal interface, if studied properly, the fetal membrane-decidua could answer many of the field’s questions regarding term labor initiation, inflammation, infection, and diseases that lead to preterm birth. To better understand the mechanism behind the labor cascade, all aspects of fetal membrane origin, as well as its cellular characteristics, need to be taken into account. Therefore, providing a global understanding of the role of fetal membranes during gestation and labor could provide insight into diagnosing and treating adverse pregnancy outcomes.

DECLARATIONS

Author contributions

Nina Truong: Reviewed the literature, and drafted the manuscript. Ramkumar Menon: Editing and reviewing the manuscript. Lauren Richardson: Conceptualization, created the figures, and draft the manuscript.

Source of funding

This manuscript is made possible by funding support from NIH/NICHD (1R01HD084532-01A1) to Ramkumar Menon. Lauren Richardson is supported by a research career development award (K12HD052023: Building Interdisciplinary Research Careers in Women's Health Program-BIRCWH; Berenson, PI) from the National Institutes of Health/Office of the Director (OD)/National Institute of Allergy and Infectious Diseases (NIAID), and Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Conflict of interest

Ramkumar Menon and Lauren Richardson are the Editorial Board Member of the journal. The article was subject to the journal’s standard procedures, with peer review handled independently of these members and their research group.

REFERENCES

  1. Mauri A, Perrini M, Mateos JM, et al. Second harmonic generation microscopy of fetal membranes under deformation: normal and altered morphology. Placenta. 2013;34(11):1020–1026.    DOI: 10.1016/j.placenta.2013.09.002
  2. Mauri A, Ehret AE, Perrini M, et al. Deformation mechanisms of human amnion: quantitative studies based on second harmonic generation microscopy. J Biomech. 2015;48(9):1606–1613.    DOI: 10.1016/j.jbiomech.2015.01.045
  3. Mauri A, Perrini M, Ehret AE, De Focatiis DSA, Mazza E. Time-dependent mechanical behavior of human amnion: macroscopic and microscopic characterization. Acta Biomater. 2015;11:314–323.    DOI: 10.1016/j.actbio.2014.09.012    PMID: 25240983
  4. Perrini M, Mauri A, Ehret AE, et al. Mechanical and microstructural investigation of the cyclic behavior of human amnion. J Biomech Eng. 2015;137(6):061010.    DOI: 10.1115/1.4030054
  5. Kumar D, Moore RM, Mercer BM, Mansour JM, Redline RW, Moore JJ. The physiology of fetal membrane weakening and rupture: insights gained from the determination of physical properties revisited. Placenta. 2016;42:59–73.    DOI: 10.1016/j.placenta.2016.03.015    PMID: 27238715
  6. Moore RM, Mansour JM, Redline RW, Mercer BM, Moore JJ. The physiology of fetal membrane rupture: insight gained from the determination of physical properties. Placenta. 2006;27(11–12):1037–1051.    DOI: 10.1016/j.placenta.2006.01.002    PMID: 16516962
  7. Richardson L, Vargas G, Brown T, et al. Redefining 3Dimensional placental membrane microarchitecture using multiphoton microscopy and optical clearing. Placenta. 2017;53:66–75.    DOI: 10.1016/j.placenta.2017.03.017
  8. Menon R, Lappas M, Zakar T. Editorial: the role of the fetal membranes in pregnancy and birth. Front Physiol. 2021;12:653084.    DOI: 10.3389/fphys.2021.653084    PMID: 33679457
  9. Zhu C, Jiang Z, Johnson GA, Burghardt RC, Bazer FW, Wu G. Nutritional and physiological regulation of water transport in the conceptus. Adv Exp Med Biol. 2022;1354:109–125.    DOI: 10.1007/978-3-030-85686-1_6
  10. Seo MJ, Lim JH, Kim DH, Bae HR. Loss of aquaporin-3 in placenta and fetal membranes induces growth restriction in mice. Dev Reprod. 2018;22(3):263–273.    DOI: 10.12717/DR.2018.22.3.263    PMID: 30324163
  11. Ducza E, Csányi A, Gáspár R. Aquaporins during pregnancy: their function and significance. Int J Mol Sci. 2017;18(12):2593.    DOI: 10.3390/ijms18122593    PMID: 29194396
  12. Kammala A, Benson M, Ganguly E, et al. Fetal membranes contribute to drug transport across the feto-maternal interface utilizing the breast cancer resistance protein (BCRP). Life. (Basel). 2022;12(2):166.    DOI: 10.3390/life12020166
  13. Ganguly E, Kammala AK, Benson M, Richardson LS, Han A, Menon R. Organic anion transporting polypeptide 2B1 in human fetal membranes: a novel gatekeeper for drug transport during pregnancy? Front Pharmacol. 2021;12:771818.    DOI: 10.3389/fphar.2021.771818    PMID: 34987396
  14. Kumar D, Springel E, Moore RM, et al. Progesterone inhibits. in vitro. fetal membrane weakening. Am J Obstet Gynecol. 2015;213(4):520.e1–520.e9.    DOI: 10.1016/j.ajog.2015.06.014
  15. Merlino A, Welsh T, Erdonmez T, et al. Nuclear progesterone receptor expression in the human fetal membranes and decidua at term before and after labor. Reprod Sci. 2009;16(4):357–363.    DOI: 10.1177/1933719108328616
  16. Lozovyy V, Richardson L, Saade G, Menon R. Progesterone receptor membrane components: key regulators of fetal membrane integrity. Biol Reprod. 2021;104(2):445–456.    DOI: 10.1093/biolre/ioaa192    PMID: 33048113
  17. Richardson LS, Taylor RN, Menon R. Reversible EMT and MET mediate amnion remodeling during pregnancy and labor. Sci Signal. 2020;13(618):eaay1486.    DOI: 10.1126/scisignal.aay1486    PMID: 32047115
  18. Fortunato SJ, Menon R, Lombardi SJ, LaFleur B. Interleukin-10 inhibition of gelatinases in fetal membranes: therapeutic implications in preterm premature rupture of membranes. Obstet Gynecol. 2001;98(2):284–288.    DOI: 10.1016/s0029-7844(01)01441-7    PMID: 11506846
  19. Hadley EE, Sheller-Miller S, Saade G, et al. Amnion epithelial cell-derived exosomes induce inflammatory changes in uterine cells. Am J Obstet Gynecol. 2018;219(5):478.e1–478478.e21.    DOI: 10.1016/j.ajog.2018.08.021
  20. Duchesne MJ, Thaler-Dao H, de Paulet AC. Prostaglandin synthesis in human placenta and fetal membranes. Prostaglandins. 1978;15(1):19–42.    DOI: 10.1016/s0090-6980(78)80003-3    PMID: 24235
  21. Keirse MJ, Turnbull AC. The fetal membranes as a possible source of amniotic fluid prostaglandins. Br J Obstet Gynaecol. 1976;83(2):146–151.    DOI: 10.1111/j.1471-0528.1976.tb00797.x    PMID: 1032816
  22. Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986–1000.    DOI: 10.1161/ATVBAHA.110.207449    PMID: 21508345
  23. Brown NL, Alvi SA, Elder MG, Bennett PR, Sullivan MH. The regulation of prostaglandin output from term intact fetal membranes by anti-inflammatory cytokines. Immunology. 2000;99(1):124–133.    DOI: 10.1046/j.1365-2567.2000.00942.x    PMID: 10651950
  24. Gibb W. The role of prostaglandins in human parturition. Ann Med. 1998;30(3):235–241.    DOI: 10.3109/07853899809005850    PMID: 9677008
  25. Mistry HD, Williams PJ. The importance of antioxidant micronutrients in pregnancy. Oxid Med Cell Longev. 2011;2011:841749.    DOI: 10.1155/2011/841749    PMID: 21918714
  26. Menon R, Richardson LS. Preterm prelabor rupture of the membranes: a disease of the fetal membranes. Semin Perinatol. 2017;41(7):409–419.    DOI: 10.1053/j.semperi.2017.07.012    PMID: 28807394
  27. Menon R, Behnia F, Polettini J, Saade GR, Campisi J, Velarde M. Placental membrane aging and HMGB1 signaling associated with human parturition. Aging. 2016;8(2):216–230.    DOI: 10.18632/aging.100891    PMID: 26851389
  28. Sheller S, Papaconstantinou J, Urrabaz-Garza R, et al. Amnion-epithelial-cell-derived exosomes demonstrate physiologic state of cell under oxidative stress. PLoS One. 2016;11(6):e0157614.    DOI: 10.1371/journal.pone.0157614
  29. Menon R. Human fetal membranes at term: Dead tissue or signalers of parturition? Placenta. 2016;44:1–5.    DOI: 10.1016/j.placenta.2016.05.013    PMID: 27452431
  30. Vora S, Abbas A, Kim CJ, et al. Nuclear factor-kappa B localization and function within intrauterine tissues from term and preterm labor and cultured fetal membranes. Reprod Biol Endocrinol. 2010;8:8.    DOI: 10.1186/1477-7827-8-8
  31. Menon R, Boldogh I, Urrabaz-Garza R, et al. Senescence of primary amniotic cells via oxidative DNA damage. PLoS One. 2013;8(12):e83416.    DOI: 10.1371/journal.pone.0083416
  32. Choltus H, Lavergne M, Belville C, et al. Occurrence of a RAGE-mediated inflammatory response in human fetal membranes. Front Physiol. 2020;11:581.    DOI: 10.3389/fphys.2020.00581
  33. Lappas M. NOD1 and NOD2 regulate proinflammatory and prolabor mediators in human fetal membranes and myometrium via nuclear factor-kappa B. Biol Reprod. 2013;89(1):14.    DOI: 10.1095/biolreprod.113.110056    PMID: 23740944
  34. Lappas M, Riley C, Lim R, et al. MAPK and AP-1 proteins are increased in term pre-labour fetal membranes overlying the cervix: regulation of enzymes involved in the degradation of fetal membranes. Placenta. 2011;32(12):1016–1025.    DOI: 10.1016/j.placenta.2011.09.011
  35. Vora S, Abbas A, Kim CJ, et al. Nuclear factor-kappa B localization and function within intrauterine tissues from term and preterm labor and cultured fetal membranes. Reprod Biol Endocrinol. 2010;8:8.    DOI: 10.1186/1477-7827-8-8
  36. Martin L, Richardson L, Menon R. Characteristics, properties, and functionality of fetal membranes: an overlooked area in the field of parturition. In: Skinner MK, ed. Encyclopedia of Reproduction. 2nd ed. Elsevier; 2018: 387–398.    DOI: 10.1016/b978-0-12-801238-3.64498-7
  37. Richardson LS, Vargas G, Brown T, et al. Discovery and characterization of human amniochorionic membrane microfractures. Am J Pathol. 2017;187(12):2821–2830.    DOI: 10.1016/j.ajpath.2017.08.019
  38. Sheller-Miller S, Urrabaz-Garza R, Saade G, Menon R. Damage-Associated molecular pattern markers HMGB1 and cell-Free fetal telomere fragments in oxidative-Stressed amnion epithelial cell-Derived exosomes. J Reprod Immunol. 2017;123:3–11.    DOI: 10.1016/j.jri.2017.08.003    PMID: 28858636
  39. Mercer BM. Preterm premature rupture of the membranes. Obstet Gynecol. 2003;101(1):178–193.    DOI: 10.1016/s0029-7844(02)02366-9    PMID: 12517665
  40. Joyce EM, Moore JJ, Sacks MS. Biomechanics of the fetal membrane prior to mechanical failure: review and implications. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S121–S127.    DOI: 10.1016/j.ejogrb.2009.02.014    PMID: 19303191
  41. Dayal S, Hong PL. Premature Rupture Of Membranes. In:. StatPearls. StatPearls Publishing; 2022.    PMID: 30422483
  42. Koh CT, Tonsomboon K, Oyen ML. Fracture toughness of human amniotic membranes. Interface Focus. 2019;9(5):20190012.    DOI: 10.1098/rsfs.2019.0012    PMID: 31485308
  43. Dars S, Malik S, Samreen I, Kazi RA. Maternal morbidity and perinatal outcome in preterm premature rupture of membranes before 37 weeks gestation. Pak J Med Sci. 2014;30(3):626–629.    DOI: 10.12669/pjms.303.4853    PMID: 24948992
  44. Medina TM, Hill DA. Preterm premature rupture of membranes: diagnosis and management. Am Fam Physician. 2006;73(4):659–664.    PMID: 16506709
  45. Dutta EH, Behnia F, Boldogh I, et al. Oxidative stress damage-associated molecular signaling pathways differentiate spontaneous preterm birth and preterm premature rupture of the membranes. Mol Hum Reprod. 2016;22(2):143–157.    DOI: 10.1093/molehr/gav074
  46. Menon R, Boldogh I, Urrabaz-Garza R, et al. Senescence of primary amniotic cells via oxidative DNA damage. PLoS One. 2013;8(12):e83416.    DOI: 10.1371/journal.pone.0083416
  47. Menon R. Oxidative stress damage as a detrimental factor in preterm birth pathology. Front Immunol. 2014;5:567.    DOI: 10.3389/fimmu.2014.00567    PMID: 25429290
  48. Bredeson S, Papaconstantinou J, Deford JH, et al. HMGB1 promotes a p38MAPK associated non-infectious inflammatory response pathway in human fetal membranes. PLoS One. 2014;9(12):e113799.    DOI: 10.1371/journal.pone.0113799
  49. Menon R, Boldogh I, Hawkins HK, et al. Histological evidence of oxidative stress and premature senescence in preterm premature rupture of the human fetal membranes recapitulated in vitro. Am J Pathol. 2014;184(6):1740–1751.    DOI: 10.1016/j.ajpath.2014.02.011
  50. Richardson L, Vargas G, Brown T, et al. Redefining 3Dimensional placental membrane microarchitecture using multiphoton microscopy and optical clearing. Placenta. 2017;53:66–75.    DOI: 10.1016/j.placenta.2017.03.017
  51. Gibbs RS, Duff P. Progress in pathogenesis and management of clinical intraamniotic infection. Am J Obstet Gynecol. 1991;164(5):1317–1326.    DOI: 10.1016/0002-9378(91)90707-x    PMID: 2035575
  52. Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol. 1993;36(4):795–808.    DOI: 10.1097/00003081-199312000-00004    PMID: 8293582
  53. Yoon BH, Romero R, Moon JB, et al. Clinical significance of intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2001;185(5):1130–1136.    DOI: 10.1067/mob.2001.117680
  54. Masucci MT, Minopoli M, Del Vecchio S, Carriero MV. The emerging role of neutrophil extracellular traps (NETs) in tumor progression and metastasis. Front Immunol. 2020;11:1749.    DOI: 10.3389/fimmu.2020.01749    PMID: 33042107
  55. Goulopoulou S, Davidge ST. Molecular mechanisms of maternal vascular dysfunction in preeclampsia. Trends Mol Med. 2015;21(2):88–97.    DOI: 10.1016/j.molmed.2014.11.009    PMID: 25541377
  56. Craige SM, Kant S, Keaney JF Jr. Reactive oxygen species in endothelial function - from disease to adaptation-. Circ J. 2015;79(6):1145–1155.    DOI: 10.1253/circj.CJ-15-0464    PMID: 25986771
  57. Veiga ECA, Korkes HA, Salomão KB, Cavalli RC. Association of LEPTIN and other inflammatory markers with preeclampsia: a systematic review. Front Pharmacol. 2022;13:966400.    DOI: 10.3389/fphar.2022.966400    PMID: 36034841
  58. Roberts JM, Lain KY. Recent insights into the pathogenesis of pre-eclampsia. Placenta. 2002;23(5):359–372.    DOI: 10.1053/plac.2002.0819    PMID: 12061851
  59. Litwiniuk M, Grzela T. Amniotic membrane: new concepts for an old dressing. Wound Repair Regen. 2014;22(4):451–456.    DOI: 10.1111/wrr.12188    PMID: 25041617
  60. Fuchs F, Riis P. Antenatal sex determination. Nature. 1956;177(4503):330.    DOI: 10.1038/177330a0    PMID: 13297032
  61. Simpson JL, Elias S. Isolating fetal cells in maternal circulation for prenatal diagnosis. Prenat Diagn. 1994;14(13):1229–1242.    DOI: 10.1002/pd.1970141308    PMID: 7617569