Crucial role of corticotropin-releasing hormone, corticotropin-releasing hormone-binding protein, mir-200c, and mir-181a in preterm delivery: A case-control study

Abstract

Background: Preterm birth before 37th wk of gestation is called premature birth. Corticotropin-releasing hormone (CRH) and CRH-binding protein (BP) act on various maternal and fetal tissues during pregnancy, such as the myometrium, which regulates the transition from the dormant phase of the uterus to the active phase. Studies have shown that mir-200c and mir-181a interact with CRH and CRH-BP.


Objective: The present study aimed to investigate the expression of mir-200c, mir-181a, CRH, and CRH-BP in women with a history of preterm birth.


Materials and Methods: In this case-control study, the gene expression level of mir-200c, mir-181a, CRH, and CRH-BP in placental tissue samples obtained from 48 women with a history of preterm labor was assessed in the Mojibian hospital of Yazd, Iran, from January to March, 2023. Differences between mir-200c, mir-181a CRH, and CRH-BP gene expressions among cases and controls were assessed.


Results: The outcomes indicated that the expression of CRH increased with going on to the regular parturition time (p < 0.001). While outcomes indicated, CRH-BP decreased with going on to the regular parturition time (p < 0.001). In addition, the results showed that the expression of mir-181a increased and mir-200c decreased with approaching the normal delivery time (p < 0.001).


Conclusion: In conclusion, the expressions of mir-200c, mir-181a, CRH, and CRH-BP were dissimilar in different weeks of gestation. It could be proposed to use mir-200c, mir-181a, CRH, and CRH-BP as biomarkers to weigh the exact delivery time, which could minimize the side effects of preterm labor for the mother and fetus.


Key words: CRH, CRH-BP, mir-200c, mir-181a, Preterm labor.

References
[1] Walani SR. Global burden of preterm birth. Int J Gynecol Obstet 2020; 150: 31-33.

[2] Waitzman NJ, Jalali A, Grosse SD. Preterm birth lifetime costs in the United States in 2016: An update. Semin Perinatol 2021; 45: 151390.

[3] Chawanpaiboon S, Vogel JP, Moller A-B, Lumbiganon P, Petzold M, Hogan D, et al. Global, regional, and national estimates of levels of preterm birth in 2014: A systematic review and modelling analysis. Lancet Glob Health 2019; 7: e37–e46.

[4] Bekkar B, Pacheco S, Basu R, DeNicola N. Association of air pollution and heat exposure with preterm birth, low birth weight, and stillbirth in the US: A systematic review. JAMA Netw Open 2020; 3: e208243.

[5] Fettweis JM, Serrano MG, Brooks JP, Edwards DJ, Girerd PH, Parikh HI, et al. The vaginal microbiome and preterm birth. Nat Med 2019; 25: 1012–1021.

[6] Yang H, Ma Q, Wang Y, Tang Z. Clinical application of exosomes and circulating microRNAs in the diagnosis of pregnancy complications and foetal abnormalities. J Transl Med 2020; 18: 32.

[7] Nazari M, Babakhanzadeh E, Aghaei Zarch SM, Talebi M, Narimani N, Dargahi M, et al. Upregulation of the RNF8 gene can predict the presence of sperm in azoospermic individuals. Clin Exp Reprod Med 2020; 47: 61–67.

[8] Babakhanzadeh E, Danaei H, Abedinzadeh M, Ashrafzadeh HR, Ghasemi N. Association of miR-146a and miR196a2 genotype with susceptibility to idiopathic recurrent pregnancy loss in Iranian women: A case-control study. Int J Reprod BioMed 2021; 19: 725–732.

[9] Williams KC, Renthal NE, Gerard RD, Mendelson CR. The microRNA (miR)-199a/214 cluster mediates opposing effects of progesterone and estrogen on uterine contractility during pregnancy and labor. Mol Endocrinol 2012; 26: 1857–1867.

[10] Renthal NE, Williams KC, Mendelson CR. MicroRNAsmediators of myometrial contractility during pregnancy and labour. Nat Rev Endocrinol 2013; 9: 391–401.

[11] Bidarimath M, Khalaj K, Wessels JM, Tayade C. MicroRNAs, immune cells and pregnancy. Cell Mol Immunol 2014; 11: 538–547.

[12] Alipourfard I, Khorshidian A, Babakhanzadeh E, Nazari M. Susceptibility to azoospermia by haplotype analysis of protamine 1 and protamine 2 variants. Human Gene 2023; 37: 201200.

[13] Thomson M. The physiological roles of placental corticotropin releasing hormone in pregnancy and childbirth. J Physiol Biochem 2013; 69: 559–573.

[14] Grammatopoulos DK. The role of CRH receptors and their agonists in myometrial contractility and quiescence during pregnancy and labour. Front Biosci 2007; 12: 561–571.

[15] Khodadadian A, Varghaiyan Y, Babakhanzadeh E, Alipourfard I, Haghi-Daredeh S, Ghobadi A, et al. Fertility preservation in women with ovarian cancer: Finding new pathways: A case-control study. Int J Reprod BioMed 2021; 19: 157–166.

[16] Kropach N, Holtzman D, Vitner D, Bar J, Chen A, Kovo M. The effect of magnesium sulfate on the placental corticotropin-releasing factor (CRF) and CRF binding protein mRNA expression in perfused human placental cotyledon. J Matern Fetal Neonatal Med 2016; 29: 376– 379.

[17] Vrachnis N, Malamas FM, Sifakis S, Tsikouras P, Iliodromiti Z. Immune aspects and myometrial actions of progesterone and CRH in labor. Clin Dev Immunol 2012; 2012: 937618.

[18] Scholten RH, Moller P, Andersen ZJ, Dehlendorff C, Khan J, Brandt J, et al. Telomere length in newborns is associated with exposure to low levels of air pollution during pregnancy. Environ Int 2021; 146: 106202.

[19] Parvez S, Gerona RR, Proctor C, Friesen M, Ashby JL, Reiter JL, et al. Glyphosate exposure in pregnancy and shortened gestational length: A prospective Indiana birth cohort study. Environ Health 2018; 17: 23.

[20] Dunn-Fletcher CE, Muglia LM, Pavlicev M, Wolf G, Sun MA, Hu Y-C, et al. Anthropoid primate–specific retroviral element THE1B controls expression of CRH in placenta and alters gestation length. PLoS Biol 2018; 16: e2006337.

[21] Klimaviciute A, Calciolari J, Bertucci E, Abelin-Tornblom S, Stjernholm-Vladic Y, Bystrom B, et al. Corticotropinreleasing hormone, its binding protein and receptors in human cervical tissue at preterm and term labor in comparison to non-pregnant state. Reprod Biol Endocrinol 2006; 4: 29.

[22] Mayor-Lynn K, Toloubeydokhti T, Cruz AC, Chegini N. Expression profile of microRNAs and mRNAs in human placentas from pregnancies complicated by preeclampsia and preterm labor. Reprod Sci 2011; 18: 46–56.

[23] You X, Gao L, Liu J, Xu C, Liu C, Li Y, et al. CRH activation of different signaling pathways results in differential calcium signaling in human pregnant myometrium before and during labor. J Clin Endocrinol Metab 2012; 97: E1851– E1861.

[24] You X, Liu J, Xu C, Liu W, Zhu X, Li Y, et al. Corticotropin-releasing hormone (CRH) promotes inflammation in human pregnant myometrium: The evidence of CRH initiating parturition? J Clin Endocrinol Metab 2014; 99: E199-E208.

[25] Zoubovsky SP, Hoseus S, Tumukuntala S, Schulkin JO, Williams MT, Vorhees CV, et al. Chronic psychosocial stress during pregnancy affects maternal behavior and neuroendocrine function and modulates hypothalamic CRH and nuclear steroid receptor expression. Transl Psychiatry 2020; 10: 6.

[26] Cathey AL, Watkins D, Rosario ZY, Vélez C, Alshawabkeh AN, Cordero JF, et al. Associations of phthalates and phthalate replacements with CRH and other hormones among pregnant women in puerto rico. J Endocr Soc 2019; 3: 1127-1149.

[27] Eick SM, Goin DE, Cushing L, DeMicco E, Smith S, Park J-S, et al. Joint effects of prenatal exposure to per-and poly-fluoroalkyl substances and psychosocial stressors on corticotropin-releasing hormone during pregnancy. J Exp Sci Environ Epidemiol 2022; 32: 27–36.