Changes in the microbiome as a factor in the development of isthmic cervical insufficiency

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

The status of a woman’s cervicovaginal microbiome may correlate with the risk of obstetric complications such as isthmic cervical insufficiency (ICI) and preterm delivery (PD). This review examined the relationship between the microbiome and ICI. The dominance of Lactobacillus crispatus and, possibly, L. gasseri in the microbiome was associated with full-term pregnancy, whereas the predominance of other Lactobacillus species and anaerobic bacteria led to the preterm rupture of membranes and PD. Notably, high levels of the antimicrobial peptide β-defensin 2, even without L. crispatus dominance, are also associated with full-term pregnancy. The analysis of the cervicovaginal and amniotic fluids of women who subsequently gave birth prematurely revealed an increase in the levels of proinflammatory cytokines, such as interleukin (IL)-2, IL-8, and IL-10. Changes in the microbiome composition and an increase in the maternal immune response lead to premature remodeling and softening of the cervix, i.e. ICI. Thus, early detection of changes in the cervicovaginal microbiome and cervicovaginal and amniotic fluids may be a prognostic marker for ICI and PD.

Full Text

Preterm birth (PTB) is commonly defined as a birth before 37 weeks of gestation. PTB is a major obstetric healthcare concern worldwide. In the United States, one in ten babies are born preterm. The incidence of PTB continues to rise annually, with an unexpectedly elevated rate in African-American females, which is approximately 50% higher than that in White or Hispanic females [1]. PTB has been associated with a wide range of maternal or fetal characteristics, such as extragenital diseases, reproductive disorders, stress factors, race, age, etc. Fuchs et al. demonstrated that a maternal age of 30–34 years was associated with the lowest risk of PTB, whereas an advanced age of 40 years and over was a strong risk factor for prematurity [2]. Moreover, vaginal microbial dysbiosis, which may result in an ascending bacterial infection from the vagina through the cervix into the uterine cavity, and cervical insufficiency (CI) may also be considered potential causes of PTB [3]. The objective of this review is to investigate the relationship between these two causes of PTB.

CI is defined by cervical shortening (<25 mm) and/or dilation (>10 mm). It is characterized by rapid, painless cervical shortening in the second or early third trimester of pregnancy, which is followed by late miscarriage or PTB. The amniotic sac may prolapse through the cervical canal, causing premature rupture of membranes (PROM). Transvaginal ultrasound (ultrasound vaginal cervicometry) is considered the most effective approach for detecting cervical insufficiency in pregnant women [4]. A lower gestational age (16–22 weeks) and a shorter cervix are associated with an increased risk (50%) of early PTB (at <32 weeks). However, this risk decreases to 15% at a later gestational age [5].

Microbiota is the set of microorganisms that colonize specific anatomical body sites. The term “microbiome” refers to both the microbiota (a set of microorganisms) and the collection of microbial genetic material in their living environment [6]. The microbiota composition may be investigated using next-generation DNA sequencing, with targeting 16S rRNA gene hypervariable regions (V1–V3 and V3–V5) as specific markers for a bacterial cell. This method offers a comprehensive qualitative and quantitative analysis of the microbiome composition [7-8]. The vaginal microbiome may be dominated by various Lactobacillus species, and several community state types (CSTs) of vaginal bacterial communities can be distinguished: L. crispatus (CST I), L. gasseri (CST II), L. iners (CST III), and L. jensenii (CST V). Another alternative is the dominance of anaerobic bacteria (Gardnerella, Atopobium, Mobiluncus, Prevotella, Streptococcus, Ureaplasma, Megasphaera, Escherichia, Shigella, etc.) and a reduction in Lactobacillus species, which contributes to bacterial vaginosis (CST IV) [9].

Menarche is associated with higher circulating estrogen levels, which leads to the proliferation of epithelial cells in the vagina and the deposition of glycogen, which is then metabolized by lactobacilli alpha-amylase to lactic acid. Lactobacillus species protect the vaginal epithelium against invasion and colonization by pathogenic and opportunistic microorganisms. The antimicrobial activity is ensured by the production of hydrogen peroxide, bacteriocins that increase the permeability of target cells, biosurfactants, and lactic acid, which reduces the pH of the medium to create unfavorable conditions for the growth and reproduction of pathogenic microorganisms. Lactic acid also facilitates the dissolution of damaged epithelial cells of the vaginal mucosa through a lysis mechanism, accompanied by glycogen production. This ultimately results in the termination of the metabolic cycle. Mucins, β-defensins, antibodies, etc. [10–11] also serve as protective agents. Lower vaginal pH (<4) is associated with higher Lactobacillus dominance, while higher pH contributes to lower Lactobacillus dominance and higher levels of anaerobic bacteria [9, 12]. Consistently high levels of estrogen, amenorrhea, and thickened endometrium during pregnancy create a favorable environment for the growth and reproduction of lactobacilli [7, 13]. PTB is associated with a highly diverse microbiome characterized by a lower dominance of Lactobacillus species (especially L. crispatus), while the vaginal microbiome of women who deliver at term is a low-diversity environment predominated by L. crispatus. Consequently, the dominance of L. crispatus is considered to be a contributing factor to full-term pregnancy [14–18]. Nevertheless, the presence of the L. crispatus-based vaginal community is not a prerequisite for full-term pregnancy and delivery at term, which are also possible with the dominance of another bacterial community [19].

In their Maternity and Microbiome (M&M) study, Elovitz et al. demonstrated that Mobiluncus curtisii/mulieris (CST IV) were most strongly associated with PTB, particularly at <34 weeks of gestation. With the low dominance of Lactobacillus spp., Mobiluncus curtisii/mulieris increase the risk of PTB, while the high dominance of Lactobacillus spp. eliminates this risk. This finding confirms a beneficial role of Lactobacillus spp. in eliminating the risk of PTB even in the presence of pathological flora [20]. A significant proportion of African-American females exhibit the low dominance of L. crispatus in their cervicovaginal microbiome, which is associated with unfavorable pregnancy outcomes. In contrast, White females demonstrate a prevalence of this Lactobacillus species. The risk of PTB in African-American females is thereby twice as high, with a higher risk of bacterial vaginosis [21–22]. However, the dominance of L. crispatus does not protect against PTB in both African-American and White females. A study of the effect of the local immune response on pregnancy outcomes, namely the role of β-defensin 2, an antimicrobial peptide that is used by immune cells to destroy phagocytized antigen, found that high levels of β-defensin 2 eliminate the risk of PTB associated with a low dominance of Lactobacillus spp. However, low levels of this antimicrobial peptide may be associated with PTB even if Lactobacillus spp. predominates [20]. During pregnancy, lower levels of β-defensin 2 may be attributed to psycho-emotional stress, and the combination of these two factors dramatically increases the risk of PTB [23].

Gerson et al. conducted a secondary analysis of the M&M study and found that the detection rate of CST IV in cervical swab specimens from women with spontaneous PTB was nearly 45%. Women with a short cervix were more susceptible to CST IV compared to other types of vaginal bacterial communities. In addition, the incidence of PTB-associated PROM was slightly higher among patients with CST IV. A short cervix and cervical CST IV have been reported to increase the risk of PTB. Furthermore, this combination was most common in African-American females [24].

Interestingly, the microbiome dominance of L. iners at 16 weeks of gestation results in preterm cervical shortening and PTB before 34 weeks of gestation, whereas the dominance of L. crispatus is associated with normal term delivery at the same gestational age. L. crispatus has been shown to dominate in White females, while African-American females exhibited the dominance of L. iners and CST IV [3]. This may be due to the fact that L. crispatus is a major source of D- and L-lactic acid, and L. iners produces only L-isomer, which has lower protective properties against pathogenic bacteria compared to D-isomer [10, 25]. Di Paola et al. showed that L. iners-dominated microbiota (among all Lactobacillus-dominated microbiotas) is most often associated with an extremely short cervix (<10 mm) and considered a risk factor for cervical remodeling and CI, as is the microbiota dominated by anaerobic bacteria (CST IV) [26]. Moreover, L. jensenii dominance could increase the risk of PTB, while L. gasseri may have a protective effect similar to that of L. crispatus and is associated with full-term pregnancy [27]. L. iners and G. vaginalis significantly increased cervical epithelial cell permeability, allowing for the water influx to the cervical stroma resulting in cervical softening, cervical remodeling, and PTB. In contrast, L. crispatus protects the cervical epithelial barrier against invading pathogens [28].

Vaginal dysbiosis induces an increased production of pro-inflammatory cytokines [29]. Sierra et al. demonstrated that cervicovaginal colonization by G. vaginalis results in increased mucin secretion and initiation of interleukin-6 (IL-6) synthesis in both cervicovaginal and amniotic fluids, despite the absence of ascending infection to the fetal membranes, placenta, and uterus. This suggests that pathogens have the ability to activate a local inflammatory response in the cervicovaginal space, which may result in reduced cervical elasticity, cervical remodeling, and PTB [30–31]. Women with an extremely short cervix (<15 mm) have a higher amniotic fluid concentrations of IL-6 and other pro-inflammatory mediators than those with a short cervix (15–25 mm). The amniotic fluid concentrations of IL-2 were increased in women with an extremely short cervix who ultimately delivered preterm compared to those who delivered at term [32]. Tarca et al. found that patients with a short cervix had increased concentrations of pro-inflammatory proteins in the amniotic fluid samples obtained by amniocentesis, particularly IL-8, MIP-1β, IL-6, and IL-10 at 16–22 weeks; IL-8, MIP-1β, and IL-6 at 22–26 weeks; and only IL-8 at 26–31 weeks. Consequently, a shorter cervix and a lower gestational age were associated with higher levels of pro-inflammatory proteins and the increased risk of early PTB (at less than 32 weeks). These proteins, also known as cytokines, are produced by macrophages and lymphocytes. They are chemotactic factors that attract neutrophils and other granulocytes to the site of inflammation [5]. The amniotic fluid from full-term pregnancies was found to be sterile [33].

The levels of D-lactic acid were highest in CST I (L. crispatus) communities, followed by CST V (L. jenseni) communities. Meanwhile, these communities had the lowest TIMP-1 (endogenous tissue inhibitor of matrix metalloproteinases) levels, which was associated with normal cervical length. The highest levels of L-lactic acid were associated with CST III (L. iners). The highest TIMP-1 levels were observed in CST IV (G. vaginalis) and CST III communities, which was associated with a short cervix and adverse pregnancy outcomes, such as PTB [34]. Yoo et al. showed that cervical insufficiency (>10 mm) and short cervix (<25 mm) correlated with cervicovaginal TIMP-1 and high levels of vitamin D binding protein (VDBP) and Dickkopf-related protein 3 (DDK-3). A combination of these markers appears to be a more useful predictor of PTB than each marker alone [35]. Concentrations of matrix metalloproteinase-8 (MMP-8) were higher in patients delivering preterm [36]. Thus, these concentrations may be useful as predictors of PTB.

Lower levels of L. crispatus and anaerobic dominance (Bacteroides, Fusobacteriales and Clostridiales) may be associated with PROM and PTB in the future [35]. Women delivered within 9 days at 24–28 weeks and within 5 days of PROM [36]. The recommended treatment for early PROM is currently aimed at accelerating fetal lung maturation (pulmonary surfactant production) by using glucocorticosteroids. Oral erythromycin 250 mg for 10 days is also required to prevent ascending infection [37]. However, Brown et al. found that erythromycin therapy induced vaginal dysbiosis and Lactobacillus depletion, particularly in women initially colonized by Lactobacillus (dominant). This was associated with a high risk of chorioamnionitis. However, erythromycin therapy in women colonized by pathogenic bacteria decreased the diversity in pathogenic communities and improved Lactobacillus dominance. Thus, there are two groups of women: those for whom erythromycin therapy is detrimental and those for whom it is potentially beneficial [16].

The above data support the hypothesis that woman's vaginal microbiome may correlate with the risk of obstetric complications such as CI or preterm birth. L. crispatus protects the cervical and vaginal epithelial barrier, which is associated with delivery at term, while the other vaginal communities are associated with the risk of CI and PTB. Activation of the maternal immune response to pathogens with increased levels of pro-inflammatory cytokines in the cervicovaginal and amniotic fluids is another risk factor for CI and PTB. However, the prevention and treatment strategies for CI and PTB should be further investigated.

ADDITIONAL INFO

Authors’ contribution. All authors made a substantial contribution to the conception of the work, acquisition, analysis, interpretation of data for the work, drafting and revising the work, final approval of the version to be published and agree to be accountable for all aspects of the work.

Funding source. This study was not supported by any external sources of funding.

Competing interests. The authors declares that there are no obvious and potential conflicts of interest associated with the publication of this article.

×

About the authors

Kamil R. Bakhtiyarov

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Author for correspondence.
Email: doctorbah@mail.ru
ORCID iD: 0000-0002-3176-5589

MD, Dr. Sci. (Medicine), Professor

Russian Federation, Moscow

Afet S. Abdulaeva

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Email: abdulaeva.a.00@gmail.com
ORCID iD: 0000-0001-6680-6400

student

Russian Federation, Moscow

Makka B. Bimurzayeva

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Email: bimakka@mail.ru
ORCID iD: 0000-0002-3065-0755

student

Russian Federation, Moscow

Daria V. Korolyova

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Email: dashak1504@gmail.com
ORCID iD: 0009-0009-9093-2103

student

Russian Federation, Moscow

Polina I. Kuzmina

I.M. Sechenov First Moscow State Medical University (Sechenov University)

Email: kuzmina_p_i@student.sechenov.ru
ORCID iD: 0009-0004-0098-1774

student

Russian Federation, Moscow

References

  1. Preterm Birth. Maternal and Infant Health. Reproductive Health. [Internet]. Centers for Disease Control and Prevention, 2020. [cited 2024 Jan 02]. Available from:
  2. https://www.cdc.gov/reproductivehealth/maternalinfanthealth/pretermbirth.htm
  3. Fuchs F, Monet B, Ducruet T, Chaillet N, Audibert F. Effect of maternal age on the risk of preterm birth: A large cohort study. PLoS One. 2018;13(1):e0191002. doi: 10.1371/journal.pone.0191002
  4. Kindinger LM, Bennett PhR, Lee YS, et al. The interaction between vaginal microbiota, cervical length, and vaginal progesterone treatment for preterm birth risk. Microbiome. 2017;5(1):6. doi: 10.1186/s40168-016-0223-9
  5. Russian Society of Obstetricians and Gynecologists. Klinicheskie rekomendatsii «Istmiko-tservikal'naya nedostatochnost' ― 2021–2022–2023». Approved by the Ministry of Health of the Russian Federation. 2021. (In Russ).
  6. Tarca AL, Fitzgerald W, Chaemsaithong P, et al. The cytokine network in women with an asymptomatic short cervix and the risk of preterm delivery. Am J Reprod Immunol. 2017;78(3):e12686. doi: 10.1111/aji.12686
  7. The Integrative Human Microbiome Project. Nature. 2019;569(7758):641–648. doi: 10.1038/s41586-019-1238-8
  8. Bayar E, Bennett PhR, Chan D, Sykes L, MacIntyre DA. The pregnancy microbiome and preterm birth. Semin Immunopathol. 2020;42(4):487–499. doi: 10.1007/s00281-020-00817-w
  9. Conlan S, Kong HH, Segre JA. Species-level analysis of DNA sequence data from the NIH Human Microbiome Project. PLoS One. 2012;7(10):e47075. doi: 10.1371/journal.pone.0047075
  10. Ravel J, Gajer P, Abdo Z, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A. 2011;108 Suppl. 1:4680–4687. doi: 10.1073/pnas.1002611107
  11. Amabebe E, Anumba DOC. The Vaginal Microenvironment: The Physiologic Role of Lactobacilli. Front Med (Lausanne). 2018;5:181. doi: 10.3389/fmed.2018.00181
  12. Wang S, Wang Q, Yang E, et al. Antimicrobial Compounds Produced by Vaginal Lactobacillus crispatus Are Able to Strongly Inhibit Candida albicans Growth, Hyphal Formation and Regulate Virulence-related Gene Expressions. Front Microbiol. 2017;8:P564. doi: 10.3389/fmicb.2017.00564
  13. Nazarova VV, Shipitsina EV, Shalepo KV, Savicheva AM. Bacterial communities forming the vaginal micro-ecosystem in nom and bacterial vaginosis. Journal of Obstetrics and Women Diseases. 2017;66(6):30–43. doi: 10.17816/JOWD66630-43
  14. Odogwu NM, Onebunne CA, Chen J, et al. Lactobacillus crispatus thrives in pregnancy hormonal milieu in a Nigerian patient cohort. Sci Rep. 2021;11(1):18152. doi: 10.1038/s41598-021-96339-y
  15. Hočevar K, Maver A, Vidmar Šimic M, et al. Vaginal Microbiome Signature Is Associated With Spontaneous Preterm Delivery. Front Med (Lausanne). 2019;6:201. doi: 10.3389/fmed.2019.00201
  16. MacIntyre DA, Chandiramani M, Lee YS, et al. The vaginal microbiome during pregnancy and the postpartum period in a European population. Sci Rep. 2015;5:8988. doi: 10.1038/srep08988
  17. Brown R, Chan D, Terzidou V, et al. Prospective observational study of vaginal microbiota pre- and post-rescue cervical cerclage. BJOG. 2019;126(7):916–925. doi: 10.1111/1471-0528.15600
  18. Dols JAM, Molenaar D, van der Helm J, et al. Molecular assessment of bacterial vaginosis by Lactobacillus abundance and species diversity. BMC Infect Dis. 2016;16:180. doi: 10.1186/s12879-016-1513-3
  19. DiGiulio DB, Сallahan BJ, McMurdie PJ, et al. Temporal and spatial variation of the human microbiota during pregnancy. Proc Natl Acad Sci U S A. 2015;112(35):11060–11065. doi: 10.1073/pnas.1502875112
  20. Stout MJ, Zhou Y, Wylie KM, et al. Early pregnancy vaginal microbiome trends and preterm birth. Am J Obstet Gynecol. 2017;217(3):356.e1–356.e18. doi: 10.1016/j.ajog.2017.05.030
  21. Elovitz MA, Gajer P, Riis V, et al. Cervicovaginal microbiota and local immune response modulate the risk of spontaneous preterm delivery. Nat Commun. 2019;10(1):1305. doi: 10.1038/s41467-019-09285-9
  22. Fettweis JM, Serrano MG, Brooks JP, et al. The vaginal microbiome and preterm birth. Nat Med. 2019;25(6):1012–1021. doi: 10.1038/s41591-019-0450-2
  23. Callahan BJ, DiGiulio DB, Goltsman DSA, et al. Replication and refinement of a vaginal microbial signature of preterm birth in two racially distinct cohorts of US women. Proc Natl Acad Sci U S A. 2017;114(37):9966–9971. doi: 10.1073/pnas.1705899114
  24. Burris HH, Riis VM, Schmidt I, et al. Maternal stress, low cervicovaginal β-defensin, and spontaneous preterm birth. Am J Obstet Gynecol MFM. 2020;2(2):100092. doi: 10.1016/j.ajogmf.2020.100092
  25. Gerson KD, McCarthy C, Elovitz MA, et al. Cervicovaginal microbial communities deficient in Lactobacillus species are associated with second trimester short cervix. Am J Obstet Gynecol. 2020;222(5):491.e1–491.e8. doi: 10.1016/j.ajog.2019.11.1283
  26. Abdelmaksoud AA, Koparde VN, Shethet NU, et al. Comparison of Lactobacillus crispatus isolates from Lactobacillus-dominated vaginal microbiomes with isolates from microbiomes containing bacterial vaginosis-associated bacteria. Microbiology (Reading). 2016;162(3):466–475. doi: 10.1099/mic.0.000238
  27. Di Paola M, Seravalli V, Paccosi S, et al. Identification of Vaginal Microbial Communities Associated with Extreme Cervical Shortening in Pregnant Women. J Clin Med. 2020;9(11):3621. doi: 10.3390/jcm9113621
  28. Stafford GP, Parker JL, Amabebe E, et al. Spontaneous Preterm Birth Is Associated with Differential Expression of Vaginal Metabolites by Lactobacilli-Dominated Microflora. Front Physiol. 2017;8:615. doi: 10.3389/fphys.2017.00615
  29. Anton L, Sierra L-J, DeVine A, et al. Common Cervicovaginal Microbial Supernatants Alter Cervical Epithelial Function: Mechanisms by Which Lactobacillus crispatus Contributes to Cervical Health. Front Microbiol. 2018;9:2181. doi: 10.3389/fmicb.2018.02181
  30. Campisciano G, Zanotta N, Licastro D, De Seta F, Comar M. In vivo microbiome and associated immune markers: New insights into the pathogenesis of vaginal dysbiosis. Sci Rep. 2018;8:2307. doi: 10.1038/s41598-018-20649-x
  31. Sierra L.-J, Brown AG, Barila GO, et al. Colonization of the cervicovaginal space with Gardnerella vaginalis leads to local inflammation and cervical remodeling in pregnant mice. PLoS One. 2018;13(1):e0191524. doi: 10.1371/journal.pone.0191524
  32. Kim YM, Park KH, Park H, et al. Complement C3a, But Not C5a, Levels in Amniotic Fluid Are Associated with Intra-amniotic Infection and/or Inflammation and Preterm Delivery in Women with Cervical Insufficiency or an Asymptomatic Short Cervix (≤ 25 mm). J Korean Med Sci. 2018;33(35):e220. doi: 10.3346/jkms.2018.33.e220
  33. Galaz J, Romero R, Xu Y, et al. Cellular immune responses in amniotic fluid of women with a sonographic short cervix. J Perinat Med. 2020. Vol. 48, N. 7. P. 665–676. doi: 10.1515/jpm-2020-0037
  34. Lim ES, Rodriguez C, Holtz LR. Amniotic fluid from healthy term pregnancies does not harbor a detectable microbial community. Microbiome. 2018;6(1):87. doi: 10.1186/s40168-018-0475-7
  35. Witkin SS, Moron AF, Ridenhour BJ, et al. Vaginal Biomarkers That Predict Cervical Length and Dominant Bacteria in the Vaginal Microbiomes of Pregnant Women. mBio. 2019;10(5):e02242-19. doi: 10.1128/mBio.02242-19
  36. Yoo H-N, Park KH, Jung EY, et al. Non-invasive prediction of preterm birth in women with cervical insufficiency or an asymptomatic short cervix (≤25 mm) by measurement of biomarkers in the cervicovaginal fluid. PLoS One. 2017;12(7):e0180878. doi: 10.1371/journal.pone.0180878
  37. Dymanowska-Dyjak I, Stupak A, Kondracka A, et al. Elastography and Metalloproteinases in Patients at High Risk of Preterm Labor. J Clin Med. 2021;10(17):3886. doi: 10.3390/jcm10173886
  38. Brown RG, Marchesi JR, Lee YS, et al. Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med. 2018;16(1):9. doi: 10.1186/s12916-017-0999-x
  39. Peaceman AM, Lai Y, Rouse DJ, et al. Length of latency with preterm premature rupture of membranes before 32 weeks’ gestation. Am J Perinatol. 2015;32(1):57–62. doi: 10.1055/s-0034-1373846
  40. Preterm labour and birth. NICE guideline [NG25] [Internet]. Recommendations. National Institute for Health and Care Excellence, 20 November 2015. [cited 2024 Jan 4]. Available from: https://www.nice.org.uk/guidance/ng25/chapter/Recommendations#intrapartum-antibiotics

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2024 Eco-Vector



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ:
ПИ № ФС 77 - 86335 от 11.12.2023 г.  
СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ:
ЭЛ № ФС 77 - 80633 от 15.03.2021 г.



This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies