The Genetic Architecture of Cervical Length is Shared with Spontaneous Preterm Birth Risk ========================================================================================= * Hope M. Wolf * Bradley T. Webb * Jerome F. Strauss III * Adi L. Tarca * Roberto Romero * Sonia S. Hassan * Shawn J. Latendresse * Tinnakorn Chaiworapongsa * Stanley Berry * Nardhy Gomez-Lopez * Piya Chaemsaithong * Timothy P. York ## ABSTRACT **Study Question** Is there a genetic contribution to the change in cervical length during pregnancy and, if so, how is this related to the known genetic contribution to pregnancy duration? **Summary Answer** Genomic analyses suggest that cervical length change across pregnancy is a polygenic trait with appreciable heritability. Bivariate genetic correlations estimated using genome-wide complex trait analysis (GCTA) indicated that a large proportion of the genes influencing cervical change across pregnancy also influenced gestational duration. **What is Known Already** Sonographic cervical length is a powerful predictor of maternal risk for spontaneous preterm birth (sPTB). Twin and family studies have established a maternal genetic heritability for sPTB ranging from 13 - 20%. However, there is no corresponding estimate for the heritability of mid-trimester cervical length, or an understanding of how genetic factors contribute to cervical changes across pregnancy. **Study Design, Size, Duration** This study was based on a prospective longitudinal cohort of (N = 5,160) Black/African American women who underwent serial sonographic examination of the uterine cervix during pregnancy and were followed until delivery. Maternal DNA extracted from whole-blood samples was genotyped via next-generation low-pass whole genome sequencing. **Participants/Materials, Settings, Methods** Repeated measurements of sonographic cervical length were collected during singleton pregnancies in 5,160 unique women and longitudinal changes in cervical length were described using latent growth models. Individual-level low-pass whole genome sequencing data from a subset of 4,423 women were used to calculate the heritability of cervical length change during pregnancy and estimate its genetic correlation with gestational duration. **Main Results and the Role of Chance** Changes in cervical length across pregnancy were found to be substantially influenced by genetic factors (*h2* = 51%). Furthermore, the genetic factors influencing cervical change were significantly correlated with genetic influences on pregnancy duration, implying a shared genetic architecture. The strongest genetic correlation was observed for the overall linear change in cervical length (*rg* = 0.982 [95% CI 0.951, 0.993]), and less so for the genetic correlation with mid-trimester values (*rg* = 0.714 [95% CI 0.419, 0.873]) and aspects of non-linear change that are most pronounced at the end of pregnancy (*rg*= -0.490 [95% CI -0.766, -0.062]). SNP-level associations were observed near genes involved in the progesterone, estrogen, and insulin signaling pathways. **Limitations, Reasons for Caution** While the study cohort was not designed to identify individual genetic variants associated with cervical length change or gestational duration, it was well powered to assess aggregate genome-wide summary statistics to estimate trait heritability, bivariate genetic correlations, and genetic enrichment of suggestive associations. To date, this study is the largest genome-wide study of preterm birth in a cohort of Black/African American women, a population that is disproportionately affected by health disparities in preterm birth and perinatal outcomes. **Wider Implications of the Findings** The significant genetic correlation between cervical length and gestational duration suggests shared causal loci between these two traits. These results raise the question of whether a large proportion of genetic loci for gestational duration exert their influence through the process of cervical remodeling. Polygenic profiling of maternal genetic liability to cervical shortening could aid in the development of clinical risk assessment tools to identify high-risk women who may benefit from more frequent cervical length screening and earlier interventions to prevent preterm delivery. **Study Funding/Completing Interest(s)** This research was supported, in part, by the Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, *Eunice Kennedy Shriver* National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS); and, in part, by federal funds from NICHD/NIH/DHHS (Contract No. HHSN275201300006C). RR has contributed to this work as part of his official duties as an employee of the United States Federal Government. ALT, NGL, and SSH were also supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health. Additional support was provided from the March of Dimes Prematurity Research Center at the University of Pennsylvania (22-FY18-812). **Trial Registration Number** N/A. Key words * cervical length * preterm birth * gestational duration * pregnancy * genetics * genome wide association study * genetic architecture ## INTRODUCTION The cervix undergoes carefully orchestrated compositional changes throughout pregnancy, transitioning from a firm structure that protects the *in utero* environment during pregnancy to a soft and pliable tissue allowing for parturition. In most pregnancies delivered at term, a gradual decrease in cervical length (CL) is observed around 30 weeks of gestation, shortening from approximately 35-45 mm to complete effacement by the first stage of labor. However, this process of cervical shortening begins earlier or happens more rapidly in some pregnancies. A short cervix is defined as a sonographic cervical length of less than 25 mm before 24 weeks of gestation, and is associated with a 6-fold increase in the risk for preterm birth (i.e., birth before 37 completed weeks)(Iams et al., 1996). The faster cervix shortens, and the earlier in pregnancy that shortening begins, the higher the risk for spontaneous preterm delivery. There is a clear pathophysiological connection between cervical dysfunction and spontaneous preterm birth, and several maternal risk factors are associated with both conditions, including maternal age, parity, pre-pregnancy BMI, race, and previous abortion (Goldenberg et al., 2008; Anum *et al*., 2010; Buck et al., 2017; Cho et al., 2017; Kandil et al., 2017; Gudicha et al., 2021; Brittain et al., 2023). A recent study showed that many maternal risk factors for preterm birth are mediated through their effects on cervical change during pregnancy (Wolf et al., 2023). Yet, whether the previously estimated genetic influences on spontaneous preterm birth (Lunde et al., 2007; Svensson et al., 2009; York et al., 2010, 2013, 2014) are related to or operate through mechanisms that affect cervical length remains unclear. While genome-wide association studies (GWAS) have identified a handful of loci associated with gestational duration (Zhang et al., 2017; Solé-Navais et al., 2023), there have been no reported heritability estimates of cervical length and only limited efforts to identify individual loci associated with cervical dysfunction (Volozonoka et al., 2020). Meta-analyses of twin and family studies have established that nearly all human complex traits are influenced by genetic factors (Claussnitzer et al., 2020). An average heritability of 45.1% was observed across 64 traits in heritability studies specifically related to female reproduction, indicating that nearly half of interindividual variation in these traits could be attributed to genetic factors (Polderman *et al*., 2015; Claussnitzer et al., 2020). Estimates for the heritability of spontaneous preterm birth range from 14 – 40% based on twin and family studies (Wadon et al., 2020) and 13 – 21% based on single nucleotide polymorphism (SNP) genotype data (York et al., 2014; Zhang et al., 2017). Heritability estimates are often correlated within functional domains, suggesting that traits related to a particular biological function share similar genetic influences. The extent to which changes in cervical length and gestational duration share a genetic architecture (i.e.,the number, location, frequency, interactions, and effect sizes of genetic variants influencing each trait) can be quantified by calculating their bivariate genetic correlation. A better understanding of the genetic and mechanistic relationships between cervical changes during pregnancy and the risk of gestational duration is needed to make progress in predicting, preventing, and treating adverse maternal and neonatal outcomes associated with premature delivery. Since there is currently no characterization of the genetic architecture of cervical length during pregnancy or how these genetic factors relate to gestational duration, this study focused on the following fundamental questions: What aspects of cervical length change across pregnancy are heritable (i.e., mid-pregnancy length, linear, and nonlinear change components)? To what extent do genetic influences on cervical change reflect a polygenic pattern of inheritance? And how much of this genetic contribution, if any, is shared with genes affecting gestational duration? Heritability estimates and the genetic correlations among traits were estimated by conducting a GWAS alongside Genome-Wide Complex Trait Analysis (GCTA) of low-pass whole-genome sequencing data in a cohort of pregnant Black/African American women with serial cervical length measurements obtained across pregnancy. Additional insight into the biological mechanisms underlying cervical change during pregnancy was gained by integrating GWAS results with functional and pathway annotations, including tissue-specific gene expression data, epigenetic marks, and chromatin accessibility. ## MATERIALS AND METHODS ### Cohort Description and Phenotyping The genotyped cohort was composed of self-identified Black/African-American women from the Detroit, Michigan area enrolled in a prospective study of pregnant women. Serial sonographic CL measurements between 8 - 40 weeks of gestation were available for 5,160 women carrying singleton pregnancies. In this study, gestational duration was evaluated as both gestational age at delivery (GAD) and spontaneous preterm birth (sPTB). GAD was measured in weeks from the first day of a woman’s last menstrual period (confirmed by ultrasound) to the day of delivery. sPTB was defined as an uninduced delivery before 37 completed weeks of gestation, including births following both spontaneous preterm labor and preterm premature rupture of membranes (PPROM). Data from patients treated with progesterone or who had cerclage (n=260) were excluded from the analysis as these interventions may alter the natural progression of cervical shortening during pregnancy. Cervical change across gestation was quantified for each pregnancy in a previous study using Latent Growth Curve Analysis (LGCA) (Wolf et al., 2022, 2023), a cross-disciplinary analytic approach that leverages repeated measurements to estimate how a trait changes within and between individuals over time. Interindividual differences in cervical change during pregnancy were described by coefficients of the growth curve corresponding to the model intercept, linear change (*i.e.*, slope), and nonlinear (*i.e.*, quadratic) change (**Figure 1**). The model intercept was centered at 17-20 weeks so that the estimated value of this coefficient would correspond to the midtrimester cervical length (MTCL). The cervical change coefficients and gestational duration measures (*i.e.*, GAD, sPTB) were used as traits in the GCTA and GWAS analyses. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F1) Figure 1. **A.** Serial measurements of CL within individuals across pregnancy. Marginal plots show distribution of CL data across gestational ages and birth outcome classes. Average trajectories of CL change during pregnancy estimated by LGCA are shown for four classes of clinical birth outcomes (extremely preterm births delivered before 28 weeks gestation, very preterm births delivered before 32 weeks gestation, late preterm births delivered before 37 weeks gestation, and full term births delivered after 37 completed weeks of gestation) are shown in color. The trajectory and rate of cervical change during pregnancy varies by gestational age at delivery. ### Ethical Approval Participants were enrolled under the protocols for Biological Markers of Disease in the Prediction of Preterm Delivery and for Preeclampsia and Intra-Uterine Growth Restriction: A Longitudinal Study (WSU IRB#110605MP2F and NICHD/NIH# OH97-CH-N067) between 2005 – 2017 at the Center for Advanced Obstetrical Care and Research (CAOCR) at Hutzel Women’s Hospital. The Institutional Review Boards of Wayne State University and the *Eunice Kennedy Shriver* National Institute of Child Health and Human Development (NICHD)/National Institutes of Health (NIH)/U.S. Department of Health and Human Services (DHHS) (Detroit, MI, USA) approved the study. All participants provided written informed consent for the collection of cervical length data and blood samples for future genetic research studies. ### Whole-Genome Sequencing and Quality Control Purified genomic DNA (gDNA) was extracted from buffy coat samples and genotyped by BGI Americas Corporation using paired-end low-pass, whole genome sequencing. Gencove, Inc. performed the sequence alignment using the GRCh37-hg19 human reference genome assembly, variant calling, phasing, and imputation based on the loimpute v0.18 algorithm for low-pass sequencing data (Li et al., 2021). A recombination map derived from the HapMap II project was used to interpolate recombination rates across all sites in the imputation reference panel. After imputation, genetic variant information was available for 65 million SNPs. The mean effective sequencing depth coverage of the sample was 1.45x. Variants with a genotyping rate of <5% (geno < 0.05); a MAF of <0.5% (MAF < 0.005); or that were found to be in extreme deviation from Hardy-Weinberg equilibrium (HWE < 0.000001) were excluded. Approximately 19.8 million single-nucleotide polymorphisms (SNPs) remained after these quality control filters. A principal component analysis (PCA) of genotypes was conducted in Plink (Purcell et al., 2007; Chang et al., 2015) based on an external reference panel of known ancestry reference (1000 Genomes Project, Phase 3) (1000 Genomes Project Consortium *et al*., 2015). Samples were excluded due to poor quality (n=2); missing data (n=15); duplicates (n=80); and population stratification concerns (n=243). In total, 4423 samples passed all quality control measures for genetic analyses (**Figure 2**). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F2) Figure 2. Quality control pipeline for genomic analyses. Purified genomic DNA from 5023 patients with available samples were sent for low-pass whole-genome sequencing. Approximately 9.8 million single-nucleotide polymorphisms (SNPs) remained after variant level filtering for genotyping rate (geno < 0.05), minor allele frequency (MAF < 0.005), and deviation from Hardy-Weinberg equilibrium (HWE < 0.000001). Samples from 4423 women remained after excluding samples for poor nucleic acid quality (n=2), missing data (n=15), duplicates (n=80), and population stratification (n=243). ### Trait Heritability and Bivariate Genetic Correlation Analysis The SNP-based heritability of cervical change coefficients was estimated using restricted maximum likelihood (REML) as implemented in GCTA version 1.92.3 (Yang et al., 2011). Covariates in the heritability analysis included sequencing depth, batch number, maternal height, pre-pregnancy BMI, parity, self-reported race/ethnicity, and the first 10 SNP-based ancestry principal components (PCs). Both self-reported race/ethnicity and ancestry principal components were included as covariates as they capture different aspects of environmental and genetic factors that may account for trait variance. SNP-based heritability was estimated for maternal height, pre-pregnancy BMI, GAD, and sPTB to compare the within-cohort estimates to SNP and twin-based heritability published in the literature. Bivariate GCTA was used to decompose observed correlations between traits into covariance components explained by genetic and environmental factors. Bivariate GCTA methods were used to estimate the genetic correlation between the cervical change coefficients and gestational duration measures. ### Genome-Wide Associations Although identification of an appreciable set of individual SNP associations was not deemed practical given the available sample size (Visscher et al., 2017; Wu et al., 2022), an exploratory analysis of genome-wide associations was conducted to gain insight into potential genetic mechanisms and biological pathways associated with cervical change and pregnancy duration. The whole genome association analysis toolset Plink (PLINK v2.00a3.3LM 64-bit Intel)(Purcell et al., 2007; Chang et al., 2015) was used to test for genetic associations with cervical change coefficients and gestational duration. PLINK dosages for the X chromosome in the all-female samples were set to levels similar to regular diploid chromosomes. Generalized linear models were used to test associations for the quantitative traits (cervical length coefficients, and GAD), while a logistic regression model was used for sPTB. Sequencing depth, batch number, self-reported race/ethnicity, height, pre-pregnancy BMI, parity, and the first 10 PCs from the PCA were variance-standardized and included as covariates. Due to the shorter extent of linkage disequilibrium (LD) in populations with African ancestry, individual SNP associations in this cohort were evaluated using the more conservative threshold for genome-wide significance (p < 1.6×10−8) based on a Bonferroni correction for an estimated 3.0 million independent LD blocks. ### Annotation of GWAS Findings The integrative web-based platform FUMA (Watanabe et al., 2017) was used to annotate variant-level summary statistics from suggestive loci (p < 1.0×10−5) associated with cervical change coefficients and gestational duration outcomes. Functional annotation and the mapping of SNPs to nearby genes was performed using the SNP2GENE function, while GENE2FUNC was used to obtain insights into putative biological mechanisms of mapped genes. Tissue specific expression patterns for the genes mapped by SNP2GENE were assessed in 53 tissue types using RNA-seq data from the Adult Genotype Tissue Expression (GTEx) v8 (GTEx Consortium, 2020). Gene-wise associations (de Leeuw et al., 2015) and Multi-marker Analysis of GenoMic Annotation (MAGMA) of the combined set of loci associated with the cervical change coefficients was used for exploratory gene-set enrichment analyses (de Leeuw et al., 2015) for a comprehensive test of genetic effects on cervical length change. ### Pre-registration Analyses were pre-registered with the Center for Open Science using the AsPredicted format ([https://osf.io/km2cg](https://osf.io/km2cg)). ## RESULTS ### SNP-Based Heritability Estimates for Cervical Length Change Cervical change during pregnancy was found to be highly heritable in the filtered sample of 4,423 women retained for genetic analyses (**Table 1**). Genetic sources explained at least 50% of inter-individual trait differences in cervical change during pregnancy (*h*2 = 0.509 [95% CI 0.327, 0.691]; *h*2 = 0.509 [95% CI 0.323, 0.695]; *h*2 = 0.507 [95% CI 0.325, 0.689]) (**Figure 3, Table S1**). The estimated heritability of GAD (*h*2 = 0.158 [95% CI -0.022, 0.338) and spontaneous preterm birth (*h*2 = 0.154 [95% CI -0.024, 0.332) were consistent with previously reported values obtained by twin and family methods (York et al., 2014; Zhang et al., 2017). For instance, the SNP-based heritability estimated for GAD in this cohort was remarkably similar to the only twin and family study (York et al., 2010) that has estimated maternal heritability for GAD in African American women (i.e., 15.8% vs. 13.8%). Heritability estimates for height (*h*2 = 0.675 [95% CI 0.506, 0.844]) and BMI (*h*2 = 0.576 [95% CI 0.400, 0.752]) were also consistent with published estimates for these traits in other cohorts (Guo et al., 2021; Yengo et al., 2022). These results show that cervical changes during pregnancy are highly heritable, and that heritability estimates from this cohort align with those reported in other studies. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F3) Figure 3. Matrix of heritability estimates and genetic and trait correlations estimated between trait pairs in the cohort. Values in the upper triangle represent observed phenotypic correlations between traits, while values in the lower triangle represent genetic correlations between traits. Values along the diagonal represent a trait’s SNP-based heritability. Note that negative genetic correlations occur when an increase in trait 1 corresponds with a decrease in trait 2. View this table: [Table 1.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T1) Table 1. Cohort Demographic Table for the n = 4423 women with low-pass whole-genome sequencing data who were included in the GCTA and GWAS genetic analyses. MTCL = observed midtrimester cervical length measurement, sCL = short cervix (defined as CL < 25 mm before 24 weeks gestation). ### Genetic Correlations between Cervical Length Change and Gestational Duration Bivariate GCTA analysis of cervical change coefficients and gestational duration suggest that genetic factors significantly contribute to the observed phenotypic correlations between these traits. The observed correlations between the intercept, linear, and nonlinear change coefficients (*r*Intercept-Linear = 0.850 [95% CI 0.846, 0.854], *r*Intercept-Nonlinear = -0.658 [95% CI -0.670, -0.646], *r*Linear-Nonlinear = -0.642 [95% CI -0.654, -0.629]) indicated that a shorter midtrimester cervical length (i.e., intercept) was associated with a more rapid rate of cervical shortening that begins earlier in pregnancy (**Figure 3**). There was significant overlap between the genetic factors influencing cervical change coefficients and gestational age at delivery (*rg*, Intercept-GAD = 0.714 [95% CI 0.419, 0.873]; *rg*, Linear-GAD= 0.982 [95% CI 0.951, 0.993]; *rg*, Nonlinear-GAD = -0.490 [95% CI -0.766, -0.062] (**Figure 3, Table S2**). Moreover, the genetic correlation between measures of gestational duration, GAD and sPTB (*rg*, GAD-sPTB = -0.600 [95% CI -1.114, -0.086]), was similar to another reported estimate (Solé-Navais et al., 2023). These results confirm a genetic correlation between changes in cervical length and gestational age at delivery, suggesting they share underlying biological mechanisms. ### Genome-Wide Associations with Gestational Duration An exploratory genome-wide analysis was conducted to identify potential genetic mechanisms associated with gestational duration. Two variants were found to be significantly associated with GAD using a genome-wide significance threshold of 1.6×10−8 (**Figure 4A**). SNPs rs75296056 and rs185443461 on Chromosome 18 (p = 1.85×10−9) are located near the gene encoding the Ras Like Without CAAX 2 (RIT2) protein. RIT2 is a calmodulin-binding GTPase that belongs to the RAS superfamily, and is thought to play a role in cellular signaling, calcium dynamics, dopamine trafficking, and autophagy (Tebar et al., 2020; Obergasteiger et al., 2023). While the relationship between RIT2 and gestational duration is not clear, it may promote the maintenance of pregnancy by regulating the autophagy-lysosomal pathway, a cellular process of protein degradation that provides energy and nutrients to maintain cellular homeostasis under stressful environmental conditions (Obergasteiger et al., 2023). Autophagic activity is essential to the processes of embryonic development, implantation, and placental development in the hypoxic and low-nutrient intrauterine environment of early pregnancy. Dysregulation of placental autophagy has been linked to several pregnancy complications, including miscarriage, gestational hypertension, gestational obesity, gestational diabetes, intrauterine growth restriction, and premature birth (Obergasteiger et al., 2023; Zhou et al., 2024). In total, 4,410 candidate SNPs, 400 independent lead SNPs, 329 genomic risk loci, and 950 mapped genes were identified using a “suggestive” association threshold of 1×10−5 (**Figure 4A**). The QQ plots for the GWAS and gene-based testing (**Figure S1**) show an acceptable level of genomic inflation (λSNP = 1.32). These results are consistent with a polygenic architecture, wherein many genes each contribute a small amount to interindividual variation in gestational duration. Genome-wide analysis in this cohort identified two novel loci significantly associated with gestational age at delivery, which were mapped to genes with functions relevant to the maintenance of pregnancy. ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F4/graphic-5.medium.gif) [](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F4/graphic-5) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F4/graphic-6.medium.gif) [](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F4/graphic-6) Figure 4 Genome Wide Associations with Cervical Change Coefficients Gestational Outcomes Manhattan plot (N = 4,423) showing the location and genome-wide association results for SNPs where the red line delineates the genome-wide significance threshold, and the blue line delineates suggestive significance threshold for cervical change coefficients, **A** intercept coefficient (I) **B** linear change coefficient (L), **C** nonlinear change coefficient (Q), and **D** gestational duration (GAD) ### Genome-Wide Associations with Cervical Change Exploratory genome-wide analyses were also performed to identify genetic associations with measures of cervical change during pregnancy. While no associations met the estimated threshold for genome-wide significance in populations with African ancestry (p < 1.6×10−8), associations with two did surpass the standard Bonferroni correction for multiple testing in GWAS (p < 5.0×10−8). Using this threshold, rs148405652 on Chromosome 4 (p = 3.85×10−8) showed a strong association with estimated mid-trimester cervical length (i.e., intercept coefficient) (**Figure 4B**). This variant maps to the Trypsin-like serine protease gene (TMPRSS11D), a Type II transmembrane serine protease that is involved in regulating many cellular processes relevant to cervical remodeling, including activation of metalloproteases, degradation of extracellular matrix components, fibroblast-to-myofibroblast differentiation, epithelial-mesenchymal transition, microvascular angiogenesis, mobilization of intracellular Ca2+, and activation latent growth factors, cytokines and hormones (Antalis et al., 2011; Menou et al., 2017). Previously known as human airway trypsin-like protease (HAT), TMPRSS11D/HAT is highly expressed in cervical and vaginal tissue samples and has been detected in the proteome of cervical-vaginal fluid(Uhlén et al., 2015; Muytjens et al., 2017; GTEx Consortium, 2020). This versatile, membrane-anchored serine protease is linked to a pro-inflammatory immune response and increased mucus production in tissues of the respiratory, lymphoid, and gastrointestinal systems (Menou et al., 2017) and has been shown to modulate the pathogenicity, spread, and tissue-specificity of viral infections (Yamaoka et al., 1998). TMPRSS11D/HAT may therefore play a crucial role in supporting the adaptive immune and innate barrier functions of mucosal surfaces in the female reproductive tract (Chokki et al., 2005; Li et al., 2014; Sheng and Hasnain, 2022). The midtrimester cervical length estimate was also associated with rs73591716 on Chromosome 10 (p = 4.63×10−8) (**Figure 4B**). This variant maps to the gene encoding calcium voltage-gated channel auxiliary subunit beta 2 (CACNB2), a subunit of a voltage-dependent calcium channel protein that controls the influx of calcium ions in cardiomyocytes, regulating the rate and contractile force of cardiomyocytes (Papa et al., 2022). This gene has been previously associated with Brugada syndrome, an inherited condition that predisposes patients to fatal cardiac arrhythmias (Antzelevitch et al., 2007; Watanabe and Minamino, 2016; Moras et al., 2023), and risk for severe psychiatric disorders (Andrade et al., 2019). Calcium ion (Ca2+) signaling plays a fundamental role in regulating uterine myometrial activity during labor and delivery, and calcium channel blockers are used clinically as tocolytic drugs to suppress uterine contractions and delay preterm labor (Gáspár and Hajagos-Tóth, 2013; Flenady et al., 2014; Wray et al., 2021). Notably, a candidate gene study of in myometrial samples from PPROM patients reported that the expression of several beta subunits of the L-type calcium channel CACNB was significantly higher in patients who delivered preterm compared to full-term deliveries (Kuć et al., 2012). An association was also observed between the linear coefficient and rs139045636 on Chromosome 3 (p = 7.67×10−8) which is located within the gene encoding Interleukin 12A + Antisense RNA (*IL-12A-AS1*) and near the gene encoding Interleukin 12A (*IL12A*) **Figure 4C**). Interleukin-12 is a pro-inflammatory cytokine produced primarily by myeloid cells, such as dendritic cells and macrophages, which induces interferon-γ (IFNγ) expression and promotes T helper 1 (Th1) differentiation, among other functions (Trinchieri, 2003; Tait Wojno *et al*., 2019). During pregnancy, there is a shift from the pro-inflammatory microenvironment associated with pregnancy establishment to a homeostatic state that persists for most of gestation to promote maternal-fetal immune tolerance (Gomez-Lopez et al., 2022). A breakdown of such maternal-fetal tolerance has therefore been associated with adverse pregnancy outcomes, including preterm labor (Hollier et al., 2004; Gomez-Lopez et al., 2022). Molecular studies suggest that IL-12 concentrations in cervical secretions are predominantly the result of local cytokine production (Castle et al., 2002). Interestingly, cervical concentrations of IL-12 have been associated with many maternal risk factors for a short cervix and preterm labor, including age, parity, genital tract infection, and a high vaginal pH (Hildesheim et al., 1999; Crowley-Nowick et al., 2000; Gravitt et al., 2003). A threshold of 1×10−5 was then used to identify “suggestive” SNPs for exploratory analyses. In total, 150 independent genomic loci containing 300 genes were identified at this threshold across all three cervical length coefficients describing cervical change during pregnancy (**Figures 4B-4D**). Of these, several suggestive SNPs were mapped to genes involved in key hormonal pathways for pregnancy; namely, Low Density Lipoprotein (LDL) Receptor Related Protein 1B (*LRP1B*) (rs141249340, p = 9.81×10−8), Relaxin 2 (*RLN2*) (rs35460272, p = 1.06×10−7), RAS like estrogen regulated growth inhibitor (RERG) (s11056394, p = 7.45×10−7), the Progesterone Receptor (*PGR*) (rs147395300, p = 7.73×10−7), Growth Regulating Estrogen Receptor Binding 1 (*GREB1*) (rs13431344, p = 3.64×10−6), and pregnancy-associated plasma protein A (*PAPPA*) (rs7865534, p = 7.14×10−6). Suggestive associations were also identified between cervical change coefficients and several proteins involved in connective tissue organization, including SPARC-Related Modular Calcium-Binding Protein 2 (SMOC2)(rs571424410, p = 5.36×10−8), ADAM metallopeptidase with thrombospondin type 1(ADAMTS1)(rs113157601, p = 3.14×10−8), Lysyl Oxidase Like 3 (*LOXL3*) (rs6546907, p = 3.46×10−7), Matrix Metallopeptidase 17 (*MMP17*) (rs74495243, p = 8.92×10−6), Fibulin 1 (*FBLN1*) (rs115293381, p = 9.37×10−6), and fibrillar collagens *COL3A1* and *COL5A2* (rs9776810, p = 9.74×10−6). ### Functional Annotation and Gene-Set Enrichment Results Functional annotations and enrichment analyses were performed on the variants associated with cervical change. Genes mapped to suggestive hits showed downregulated expression in GTEx v8 endocervical tissue (**Figure S2**). Gene-set enrichment profiling of all genes mapped to suggestive SNPs for cervical change resulted in significant enrichment of molecular and functional domains based on a false discovery rate (FDR) of 5% **(Tables S3-S11**). Identified Kegg and Gene Ontology (GO) pathways corresponded to biological processes and molecular functions related to growth factors, energy metabolism, hormonal signaling, immune response, and inflammation (**Tables S7-S10**), including the Interleukin-2 pathways (p = 6.56×10−6) and the Interleukin-20 (p = 9.12×10−6), Interferon (p = 1.97×10−5), and Vascular Endothelial Growth Factor (*VEGFA*, p = 4.88×10−4) signaling pathways. Glucose and lipid metabolism (p = 1.57×10−4) are vital for fetal development and energy homeostasis during pregnancy, and alterations in these metabolic pathways could negatively impact fetal growth and development, affecting gestational length (Parrettini et al., 2020; Jovandaric et al., 2023). Estrogen signaling (p = 1.91×10−4) plays a significant role in preparing the body for labor and delivery by promoting the production of prostaglandins, lipid compounds that mediate cervical ripening and stimulate myometrial contractions to initiate labor (Socha et al., 2022). Likewise, activation of immune cells and the release of cytokines contributes to an inflammatory response in the uterus that is conducive to the onset of labor (Yockey and Iwasaki, 2018). Differentially expressed gene sets from 54 tissue types (GTEx v8) suggested that many of the mapped genes are differentially expressed in endocervical cells (**Figure S2**). In contrast to the ectocervix, which protrudes into the vagina and is covered by stratified squamous epithelial cells, the endocervix is composed of the columnar epithelial cells that line the cervical canal and internal os and may be influenced by molecular and hormonal signaling from the chorioamniotic membranes (Mendelson et al., 2017; Yellon, 2019; Socha et al., 2022). Although these results suggest that many of the mapped genes could be expressed in endocervical tissue, the differential regulation results should be interpreted with caution as the 10 cervical tissue samples included in GTEx V8 were collected from non-pregnant donors, many of whom were likely postmenopausal, and thus do not reflect gene expression changes that occur during pregnancy. ## DISCUSSION Proper cervical length remodeling throughout pregnancy is integral to successfully maintaining a pregnancy and preparing for delivery. While there are many suspected factors that can precipitate early labor (Romero et al., 2014; Strauss et al., 2018), pathological shortening and effacement of the cervix during pregnancy provides a clear mechanism of preterm birth (Iams et al., 1996; Myers et al., 2015). This study was designed to provide the first assessment of the genetic architecture of cervical length and quantify its genetic relationship to gestational duration. Low-pass sequencing experiments allowed for the estimation of the number, effect size, and population frequency of genetic variants essential for heritability studies of cervical length and its rate of change across pregnancy. The findings of the current study have contributed two fundamental insights. First, genetic factors accounted for approximately 50% of inter-individual differences in cervical change during pregnancy. These heritability estimates are consistent with a highly polygenic, complex trait influenced by both genetic and environmental factors (Romero et al., 2014; Strauss et al., 2018). The estimated heritability of gestational duration outcomes and other biometric measurements were consistent with published estimates for these traits obtained by twin and family and SNP-based estimates in other studies (York et al., 2014; Zhang et al., 2017; Guo et al., 2021; Yengo et al., 2022). Specifically, the estimated SNP-based maternal heritability of GAD from this study of self-identified African American women (*h2* = 15.8%) was similar to the only estimate available from an African American twin and family cohort (*h2* = 13.8%) (York et al., 2010). The tendency for SNP-based heritability estimates to be lower than their twin-based counterparts (Friedman et al., 2021) was not observed and preliminarily suggests that common genetic alleles, in contrast to rare or structural variants, accounted for the majority of genetic variation in gestational age at birth. Second, the robust genetic correlations estimated among cervical change and gestational duration traits suggested that a large proportion of the genetic factors contributing to cervical change also influenced gestational duration. Estimates of the genetic correlation along with the trait-specific heritability can be used to provide an approximation of the observed bivariate trait correlations expected to be due to correlated genetic factors (Plomin et al., 2012). For example, the observed trait correlation between GAD and the MTCL (i.e., intercept) measures was 0.232 of which 87.1% was estimated to be due to shared genetic factors. Similarly, the GAD-linear (*r*GAD-Linear = 0.246) and GAD-nonlinear (*r*GAD-Nonlinear = -0.148) trait correlations were estimated to be almost entirely due to genetic factors, near 100% and 93.9%, respectively. A meta-analysis of genetic correlations across human diseases and traits confirms that a genetic correlation (*rg*) of 0.30 or higher is a reasonable expectation for two traits with a robust epidemiological association, such as cervical length and gestational duration (Bulik-Sullivan et al., 2015). It is worth noting that the replication of the GAD-sPTB genetic correlation in the current study (*rg* = -0.600 [95% CI -1.114, -0.086]), which was first described by Solé-Navais *et. al.* (*rg* = -0.62 [95% CI -0.72, -0.51]) (Solé-Navais et al., 2023), does not confirm the genetic similarity of these traits but rather indicates the loss of genetic information after dichotomizing GAD (York et al., 2013). The gene set enrichment and pathway analysis of GWAS summary statistics have shed light on biological mechanisms involved in cervical changes during pregnancy and gestational duration. Pathway enrichment analysis of suggestive variants in this cohort revealed an increased burden in molecular processes related to hormone and growth factor signaling, energy metabolism, tissue remodeling, inflammation, and immune regulation. These biological pathways have been previously implicated in the onset of labor in both premature and term birth. Genomic signals near genes involved in insulin, progesterone, and estrogen signaling, and various cytokines and growth factors like IL-2, IL-20, VEGFA, and interferon signaling were also identified. These results align with current knowledge regarding hormonal influences and responses in cervical tissue, and could offer a potential mechanistic explanation for why intervention with vaginal progesterone might help prolong gestation and prevent preterm birth in women with a short cervix. Several variants were found to be associated with cervical change and gestational duration. RLN2 is an ovarian hormone that promotes loosening of connective tissues in the pelvic ligaments and the cervix to allow for easier passage of the infant during labor and delivery (Parry and Vodstrcil, 2007). Similar to progesterone, relaxin is thought to promote pregnancy maintenance and uterine quiescence to prevent spontaneous abortions and the premature onset of labor (Goldsmith and Weiss, 2009). Although studies addressing the specific functions of RERG and GREB1 in pregnancy are limited, these proteins are thought to play a role in regulating hormone-responsive pathways in reproductive tissues (Finlin et al., 2001; Key et al., 2006; Deschênes et al., 2007; Mohammed et al., 2013). Other hormonally-responsive genes identified herein include LoxL3, which is shown to be physiologically regulated by estrogen in mice (Ozasa et al., 1981; Akins et al., 2011; Mahendroo, 2012), and FBLN1, a calcium-binding secreted glycoprotein with repeated epidermal-growth-factor-like domains that is also regulated by estrogen (Argraves et al., 1990; Clinton et al., 1996; Moll et al., 2002). A number of other genes identified herein are implicated in tissue remodeling processes in the context of pregnancy, including MMP17, a membrane-type matrix metalloproteinase that degrades components of the extracellular matrix during tissue remodeling related to embryonic development and reproduction (Yip et al., 2019). Fibulins are associated with the formation and rebuilding of extracellular matrices and elastic fibers, including connective tissue disorders in humans (de Vega et al., 2009), and FBLN1 has been associated with HPV infection leading to cervical precancerous lesions and cervical carcinoma (Hao et al., 2021). COL3A1 and COL5A2 produce alpha chains for a low abundance fibrillar collagens, and mutations in these gene are associated with Ehlers-Danlos syndrome, types I and II, which themselves are associated with an increased risk for cervical insufficiency, sPTB, and PPROM (Modi et al., 2017; Strauss et al., 2018). While its specific function during pregnancy is not understood, LRP1B has been implicated in modulating inflammatory responses and and may interact with extracellular matrix proteins or cell surface receptors to regulate cell adhesion and migration in the uterus and placenta during pregnancy (Haas et al., 2011; Sizova et al., 2023). Finally, PAPPA encodes a metalloproteinase that regulates the Insulin-like Growth Factor (IGF) pathway (Oxvig, 2015), and low serum levels of PAPPA in the first trimester have been associated with an increased risk of many adverse pregnancy outcomes, including preterm birth, low birth weight, preeclampsia, miscarriage, and stillbirth (Smith et al., 2002; Kirkegaard et al., 2010). The current study was designed to characterize several features of the genetic architecture of cervical change during pregnancy, but was not designed to identify a comprehensive set of common SNP effects. Larger sample sizes will be needed to detect robust statistical associations for individual genetic variants. This study did not address the influence of fetal genes, the vaginal microbiome, or the broader set of environmental and sociodemographic factors that may also contribute to cervical change during pregnancy. Additionally, further study is required to understand the mechanisms behind the generation of genetic correlations between these traits, which could be due to different forms of pleiotropy, linkage disequilibrium, or uncontrolled sources of bias (Solovieff et al., 2013). Previous genetic studies of preterm birth have focused on individuals of European ancestry in Scandinavia and the Americas, limiting the applicability of their findings across diverse global populations. This project is the first large genomic study of birth outcomes in a cohort of US-based women with African ancestry, who are underrepresented in genomic studies, and suffer disproportionately higher rates of sCL and sPTB. Conducting research in a high-risk and underrepresented population could contribute to more effective, personalized prevention strategies for patients from a similar genetic and sociodemographic background. Collectively, the data herein provide evidence for a substantial genetic influence on changes in cervical length throughout pregnancy. Moreover, such genetic factors are highly correlated with those influencing pregnancy duration, implying shared causal loci between these traits. By undertaking functional and pathway annotation of the observed genetic variants, we identified multiple individual genes and signaling pathways that may be implicated as determinants of cervical changes and gestational duration. Although genetic correlations have not been widely applied to birth outcomes research, they have been extensively used in the fields of psychiatric genetics and endocrinology/cardiology to predict genetic liability for correlated outcomes, enhance causal and mediation analyses, and increase statistical power for multi-trait genetic association studies(Bulik-Sullivan et al., 2015). Thus, establishing the shared genetic risk between cervical shortening and gestational duration may improve the utility of cervical length as a screening tool, provide targets for future pharmacogenetic studies, and encourage broader screening programs to identify women with a short cervix who would benefit from clinical interventions, with the ultimate goal of preventing preterm birth. ## DATA AVAILABILITY All data produced in the present study are available upon reasonable request to the authors. ## AUTHOR’S ROLES T.P.Y., J.F.S, and R.R. conceived and designed the study. H.M.W, T.P.Y., A.L.T., S.J.L., and B.T.W. contributed to analysis of the data. R.R., S.S.H., T.C., S.B., N.G.L., and P.C. acquired the data and biospecimens. T.P.Y. and H.M.W. developed the manuscript draft. J.F.S, B.T.W., A.L.T., N.G.L., and R.R. critically revised the article. All authors gave final approval of the version to be published. ## FUNDING This research was supported by the Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, *Eunice Kennedy Shriver* National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services (NICHD/NIH/DHHS) (Contract No. HHSN275201300006C). RR has contributed to this work as part of his official duties as an employee of the United States Federal Government. A.L.T., N.G.L., and S.S.H. were also supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health. Additional support was provided from the March of Dimes Prematurity Research Center at the University of Pennsylvania (22-FY18-812). ## CONFLICT OF INTEREST The funders had no influence on the data collection, analyses or conclusions of the study. There is no conflict of interest to declare. ## SUPPLEMENTAL MATERIALS View this table: [Table S1.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T2) Table S1. Heritability estimates for cervical length trajectory coefficients, and gestational duration outcomes, height, and BMI. VG represents a trait’s total genetic variance; VE represents a trait’s total environmental variance; VP represent a trait’s total phenotypic variance; h2 (VG / Vp) represents a trait’s estimated heritability, or the proportion of phenotypic variance explained by all SNPs for each trait. Literature values of SNP-based heritability are included for comparison for traits that have been studied in other cohorts. (*I*: intercept; *L*: linear term; *Q*: quadratic term). View this table: [Table S2.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T3) Table S2. Estimates for generic correlations between cervical length growth parameters and gestational duration. Note: trait 1 represents the first trait listed in the trait name (for the correlation between I & GAD, I is trait 1 and GAD is trait 2). CorP represents the correlation between the phenotypic values of the two traits. VG t1 and VG t2 represent the variation from additive genetic effects for traits 1 and 2, respectively; CGt12 represents the genetic covariance between traits 1 and 2; VE t1 and VE t2 represent the residual/environmental variance for traits 1 and 2, respectively; CEt12 represents the residual/environmental covariance between traits 1 and 2; VP t1 and VP t2 represent the total phenotypic variation traits 1 and 2, respectively; h2 (VG / Vp) represents the proportion of phenotypic variance explained by all SNPs for each trait; and rG represents the genetic correlation between trait 1 and 2. View this table: [Table S3.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T4) Table S3. Top 10 gene-set enrichment categories identified using gene-based tests of GWAS summary statistics for the intercept coefficient (I) computed by MAGMA. View this table: [Table S4.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T5) Table S4. Top 10 gene-set enrichment categories identified using gene-based tests of GWAS summary statistics for the linear change coefficient (L), computed by MAGMA. View this table: [Table S5.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T6) Table S5. Top 10 gene-set enrichment categories identified using gene-based tests of GWAS summary statistics for the nonlinear change coefficient (Q), computed by MAGMA. View this table: [Table S6.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T7) Table S6. Positional gene enrichment sets, using summary statistics GWA Studies for Cervical Change coefficients (I,L, and Q) View this table: [Table S7.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T8) Table S7. Kegg Pathway Analysis for Cervical Change coefficients (I,L, and Q) View this table: [Table S8.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T9) Table S8. Gene Ontology: Biological Processes for Cervical Change coefficients (I,L, and Q) View this table: [Table S9.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T10) Table S9. Gene Ontology: Molecular Functions for Cervical Change coefficients (I,L, and Q) View this table: [Table S10.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T11) Table S10. Gene Ontology: Cellular Components for Cervical Change coefficients (I,L, and Q) View this table: [Table S11.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T12) Table S11. Gene-set enrichment categories identified using gene-based tests of GWAS summary statistics from GWA studies of Cervical Change coefficients (I,L, and Q), computed by MAGMA. View this table: [Table S12.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/T13) Table S12. Gene-set enrichment categories identified using gene-based tests of GWAS summary statistics for GAD, computed by MAGMA. ## SUPPLEMENTAL FIGURES ![Figure S1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F5.medium.gif) [Figure S1.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F5) Figure S1. QQ-plots for GWA Studies. **A** QQ-plot for Intercept coefficient (I), **B** QQ-plot for Linear change coefficient (L), **C** QQ-plot for Nonlinear change coefficient (Q), **D** QQ-plot for GAD. ![Figure S2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/05/24/2024.05.23.24307808/F6.medium.gif) [Figure S2.](http://medrxiv.org/content/early/2024/05/24/2024.05.23.24307808/F6) Figure S2. Enriched differentially expressed gene (DEG) sets based on RNA-seq data from 54 tissue types from GTEx v8 ((GTEx Consortium, 2020). Many of the genes mapped to suggestive associations are differentially expressed in endocervical tissue, which is composed of the columnar epithelial cells that line the cervical canal and internal os. The cervical tissue samples includedGTEx V8 were collected from 10 non-pregnant donors, and do not reflect gene expression changes that occur during pregnancy. ## Footnotes * * The Perinatology Research Branch, NICHD/NIH/DHHS, has been renamed as the Pregnancy Research Branch, NICHD/NIH/DHHS. * Received May 23, 2024. * Revision received May 23, 2024. * Accepted May 23, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. 1000 Genomes Project Consortium, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA, et al. A global reference for human genetic variation. Nature 2015;526:68–74. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature15393&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26432245&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 2. Akins ML, Luby-Phelps K, Bank RA, Mahendroo M. Cervical softening during pregnancy: regulated changes in collagen cross-linking and composition of matricellular proteins in the mouse. Biol Reprod 2011;84:1053–1062. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1095/biolreprod.110.089599&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21248285&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000289728600025&link_type=ISI) 3. Andrade A, Brennecke A, Mallat S, Brown J, Gomez-Rivadeneira J, Czepiel N, Londrigan L. Genetic Associations between Voltage-Gated Calcium Channels and Psychiatric Disorders. Int J Mol Sci [Internet] 2019;20.: 4. Antalis TM, Bugge TH, Wu Q. Membrane-anchored serine proteases in health and disease. Prog Mol Biol Transl Sci 2011;99:1–50. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/B978-0-12-385504-6.00001-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21238933&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 5. Antzelevitch C, Pollevick GD, Cordeiro JM, Casis O, Sanguinetti MC, Aizawa Y, Guerchicoff A, Pfeiffer R, Oliva A, Wollnik B, et al. Loss-of-function mutations in the cardiac calcium channel underlie a new clinical entity characterized by ST-segment elevation, short QT intervals, and sudden cardiac death. Circulation 2007;115:442–449. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNpcmN1bGF0aW9uYWhhIjtzOjU6InJlc2lkIjtzOjk6IjExNS80LzQ0MiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzA1LzI0LzIwMjQuMDUuMjMuMjQzMDc4MDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 6. Anum EA, Brown HL, Strauss JF 3rd. Health disparities in risk for cervical insufficiency. Hum Reprod 2010;25:2894–2900. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/humrep/deq177&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20643692&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 7. Argraves WS, Tran H, Burgess WH, Dickerson K. Fibulin is an extracellular matrix and plasma glycoprotein with repeated domain structure. J Cell Biol 1990;111:3155–3164. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNiIjtzOjU6InJlc2lkIjtzOjEwOiIxMTEvNi8zMTU1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDUvMjQvMjAyNC4wNS4yMy4yNDMwNzgwOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 8. Brittain JJ, Wahl SE, Strauss JF 3rd, Romero R, Wolf HM, Murphy K, Cyrus JW, York TP. Prior Spontaneous or Induced Abortion Is a Risk Factor for Cervical Dysfunction in Pregnant Women: a Systematic Review and Meta-analysis. Reprod Sci [Internet] 2023;Available from: doi:10.1007/s43032-023-01170-7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s43032-023-01170-7&link_type=DOI) 9. Buck JN, Orzechowski KM, Berghella V. Racial disparities in cervical length for prediction of preterm birth in a low risk population. J Matern Fetal Neonatal Med 2017;30:1851–1854. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 10. Bulik-Sullivan B, Finucane HK, Anttila V, Gusev A, Day FR, Loh P-R, ReproGen Consortium, Psychiatric Genomics Consortium, Genetic Consortium for Anorexia Nervosa of the Wellcome Trust Case Control Consortium 3, Duncan L, et al. An atlas of genetic correlations across human diseases and traits. Nat Genet 2015;47:1236–1241. 11. Castle PE, Hildesheim A, Bowman FP, Strickler HD, Walker JL, Pustilnik T, Edwards RP, Crowley-Nowick PA. Cervical concentrations of interleukin-10 and interleukin-12 do not correlate with plasma levels. J Clin Immunol 2002;22:23–27. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1023/A:1014252402630&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11958590&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000174884800004&link_type=ISI) 12. Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 2015;4:7. 13. Chokki M, Eguchi H, Hamamura I, Mitsuhashi H, Kamimura T. Human airway trypsin-like protease induces amphiregulin release through a mechanism involving protease-activated receptor-2-mediated ERK activation and TNF alpha-converting enzyme activity in airway epithelial cells. FEBS J 2005;272:6387–6399. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1742-4658.2005.05035.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16336275&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 14. Cho SH, Park KH, Jung EY, Joo JK, Jang JA, Yoo HN. Maternal Characteristics, Short Mid-Trimester Cervical Length, and Preterm Delivery. J Korean Med Sci 2017;32:488–494. 15. Claussnitzer M, Cho JH, Collins R, Cox NJ, Dermitzakis ET, Hurles ME, Kathiresan S, Kenny EE, Lindgren CM, MacArthur DG, et al. A brief history of human disease genetics. Nature 2020;577:179–189. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-019-1879-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31915397&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 16. Clinton GM, Rougeot C, Derancourt J, Roger P, Defrenne A, Godyna S, Argraves WS, Rochefort H. Estrogens increase the expression of fibulin-1, an extracellular matrix protein secreted by human ovarian cancer cells. Proc Natl Acad Sci U S A 1996;93:316–320. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czo4OiI5My8xLzMxNiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzA1LzI0LzIwMjQuMDUuMjMuMjQzMDc4MDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 17. Crowley-Nowick PA, Ellenberg JH, Vermund SH, Douglas SD, Holland CA, Moscicki AB. Cytokine profile in genital tract secretions from female adolescents: impact of human immunodeficiency virus, human papillomavirus, and other sexually transmitted pathogens. J Infect Dis 2000;181:939–945. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/315311&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10720516&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000086344400018&link_type=ISI) 18. Deschênes J, Bourdeau V, White JH, Mader S. Regulation of GREB1 transcription by estrogen receptor alpha through a multipartite enhancer spread over 20 kb of upstream flanking sequences. J Biol Chem 2007;282:17335–17339. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyODIvMjQvMTczMzUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wNS8yNC8yMDI0LjA1LjIzLjI0MzA3ODA4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 19. Finlin BS, Gau CL, Murphy GA, Shao H, Kimel T, Seitz RS, Chiu YF, Botstein D, Brown PO, Der CJ, et al. RERG is a novel ras-related, estrogen-regulated and growth-inhibitory gene in breast cancer. J Biol Chem 2001;276:42259–42267. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyNzYvNDUvNDIyNTkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wNS8yNC8yMDI0LjA1LjIzLjI0MzA3ODA4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 20. Flenady V, Wojcieszek AM, Papatsonis DNM, Stock OM, Murray L, Jardine LA, Carbonne B. Calcium channel blockers for inhibiting preterm labour and birth. Cochrane Database Syst Rev 2014;2014:CD002255. 21. Friedman NP, Banich MT, Keller MC. Twin studies to GWAS: there and back again. Trends Cogn Sci 2021;25:855–869. 22. Gáspár R, Hajagos-Tóth J. Calcium channel blockers as tocolytics: principles of their actions, adverse effects and therapeutic combinations. Pharmaceuticals 2013;6:689–699. 23. Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. Lancet 2008;371:75–84. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(08)60074-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18177778&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000252192600033&link_type=ISI) 24. Goldsmith LT, Weiss G. Relaxin in human pregnancy. Ann N Y Acad Sci 2009;1160:130–135. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1749-6632.2008.03800.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19416173&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265652100024&link_type=ISI) 25. Gomez-Lopez N, Galaz J, Miller D, Farias-Jofre M, Liu Z, Arenas-Hernandez M, Garcia-Flores V, Shaffer Z, Greenberg JM, Theis KR, et al. The immunobiology of preterm labor and birth: intra-amniotic inflammation or breakdown of maternal-fetal homeostasis. Reproduction 2022;164:R11–R45. 26. Gravitt PE, Hildesheim A, Herrero R, Schiffman M, Sherman ME, Bratti MC, Rodriguez AC, Morera LA, Cardenas F, Bowman FP, et al. Correlates of IL-10 and IL-12 concentrations in cervical secretions. J Clin Immunol 2003;23:175–183. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1023/A:1023305827971&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12797539&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000182166900003&link_type=ISI) 27. GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 2020;369:1318–1330. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNjkvNjUwOS8xMzE4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDUvMjQvMjAyNC4wNS4yMy4yNDMwNzgwOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 28. Gudicha DW, Romero R, Kabiri D, Hernandez-Andrade E, Pacora P, Erez O, Kusanovic JP, Jung E, Paredes C, Berry SM, et al. Personalized assessment of cervical length improves prediction of spontaneous preterm birth: a standard and a percentile calculator. Am J Obstet Gynecol 2021;224:288.e1–e288.e17. Elsevier. 29. Guo J, Bakshi A, Wang Y, Jiang L, Yengo L, Goddard ME, Visscher PM, Yang J. Quantifying genetic heterogeneity between continental populations for human height and body mass index. Sci Rep 2021;11:5240. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-021-84739-z&link_type=DOI) 30. Haas J, Beer AG, Widschwendter P, Oberdanner J, Salzmann K, Sarg B, Lindner H, Herz J, Patsch JR, Marschang P. LRP1b shows restricted expression in human tissues and binds to several extracellular ligands, including fibrinogen and apoE-carrying lipoproteins. Atherosclerosis 2011;216:342–347. 31. Hao Y, Ye M, Chen X, Zhao H, Hasim A, Guo X. Discovery and validation of FBLN1 and ANT3 as potential biomarkers for early detection of cervical cancer. Cancer Cell Int 2021;21:125. 32. Hildesheim A, McShane LM, Schiffman M, Bratti MC, Rodriguez AC, Herrero R, Morera LA, Cardenas F, Saxon L, Bowman FP, et al. Cytokine and immunoglobulin concentrations in cervical secretions: reproducibility of the Weck-cel collection instrument and correlates of immune measures. J Immunol Methods 1999;225:131–143. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0022-1759(99)00037-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10365790&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000080510300014&link_type=ISI) 33. Hollier LM, Rivera MK, Henninger E, Gilstrap LC 3rd, Marshall GD Jr. T helper cell cytokine profiles in preterm labor. Am J Reprod Immunol 2004;52:192–196. 34. Iams JD, Goldenberg RL, Meis PJ, Mercer BM, Moawad A, Das A, Thom E, McNellis D, Copper RL, Johnson F, et al. The Length of the Cervix and the Risk of Spontaneous Premature Delivery. N Engl J Med 1996;334:567–573. Massachusetts Medical Society. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJM199602293340904&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8569824&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1996TW69600004&link_type=ISI) 35. Jovandaric MZ, Babic S, Raus M, Medjo B. The Importance of Metabolic and Environmental Factors in the Occurrence of Oxidative Stress during Pregnancy. Int J Mol Sci [Internet] 2023;24.: 36. Kandil M, Sanad Z, Sayyed T, Ellakwa H. Body mass index is linked to cervical length and duration of pregnancy: An observational study in low risk pregnancy. J Obstet Gynaecol 2017;37:33–37. 37. Key MD, Andres DA, Der CJ, Repasky GA. Characterization of RERG: an estrogen-regulated tumor suppressor gene. Methods Enzymol 2006;407:513–527. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0076-6879(05)07041-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16757349&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000237082800041&link_type=ISI) 38. Kirkegaard I, Uldbjerg N, Oxvig C. Biology of pregnancy-associated plasma protein-A in relation to prenatal diagnostics: an overview. Acta Obstet Gynecol Scand 2010;89:1118–1125. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3109/00016349.2010.505639&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20804336&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 39. Kuć P, Laudański P, Kowalczuk O, Chyczewski L, Laudański T. Expression of selected genes in preterm premature rupture of fetal membranes. Acta Obstet Gynecol Scand 2012;91:936–943. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22568870&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 40. Leeuw CA de, Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput Biol 2015;11:e1004219. 41. Li JH, Mazur CA, Berisa T, Pickrell JK. Low-pass sequencing increases the power of GWAS and decreases measurement error of polygenic risk scores compared to genotyping arrays. Genome Res 2021;31:529–537. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjg6IjMxLzQvNTI5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDUvMjQvMjAyNC4wNS4yMy4yNDMwNzgwOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 42. Li Y, Feng C, Wei X, Zhang J, Zan R, Zheng G, Yang X, Zhai J. Activation of protease-activated receptor-2 disrupts vaginal epithelial barrier function. Cell Biol Int 2014;38:1247–1251. 43. Lunde A, Melve KK, Gjessing HK, Skjaerven R, Irgens LM. Genetic and environmental influences on birth weight, birth length, head circumference, and gestational age by use of population-based parent-offspring data. Am J Epidemiol 2007;165:734–741. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/aje/kwk107&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17311798&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000244959600002&link_type=ISI) 44. Mahendroo M. Cervical remodeling in term and preterm birth: insights from an animal model. Reproduction 2012;143:429–438. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoicmVwcm9kIjtzOjU6InJlc2lkIjtzOjk6IjE0My80LzQyOSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzA1LzI0LzIwMjQuMDUuMjMuMjQzMDc4MDguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 45. Mendelson CR, Montalbano AP, Gao L. Fetal-to-maternal signaling in the timing of birth. J Steroid Biochem Mol Biol 2017;170:19–27. 46. Menou A, Duitman J, Flajolet P, Sallenave J-M, Mailleux AA, Crestani B. Human airway trypsin-like protease, a serine protease involved in respiratory diseases. Am J Physiol Lung Cell Mol Physiol 2017;312:L657–L668. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1152/ajplung.00509.2016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28235951&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 47. Modi BP, Teves ME, Pearson LN, Parikh HI, Chaemsaithong P, Sheth NU, York TP, Romero R, Strauss JF 3rd. Rare mutations and potentially damaging missense variants in genes encoding fibrillar collagens and proteins involved in their production are candidates for risk for preterm premature rupture of membranes. PLoS One 2017;12:e0174356. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 48. Mohammed H, D’Santos C, Serandour AA, Ali HR, Brown GD, Atkins A, Rueda OM, Holmes KA, Theodorou V, Robinson JLL, et al. Endogenous purification reveals GREB1 as a key estrogen receptor regulatory factor. Cell Rep 2013;3:342–349. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.celrep.2013.01.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23403292&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000321895200010&link_type=ISI) 49. Moll F, Katsaros D, Lazennec G, Hellio N, Roger P, Giacalone P-L, Chalbos D, Maudelonde T, Rochefort H, Pujol P. Estrogen induction and overexpression of fibulin-1C mRNA in ovarian cancer cells. Oncogene 2002;21:1097–1107. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.onc.1205171&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11850827&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000173580100013&link_type=ISI) 50. Moras E, Gandhi K, Narasimhan B, Brugada R, Brugada J, Brugada P, Krittanawong C. Genetic and Molecular Mechanisms in Brugada Syndrome. Cells [Internet] 2023;12.: 51. Muytjens CMJ, Yu Y, Diamandis EP. Discovery of Antimicrobial Peptides in Cervical-Vaginal Fluid from Healthy Nonpregnant Women via an Integrated Proteome and Peptidome Analysis. Proteomics [Internet] 2017;17.: 52. Myers KM, Feltovich H, Mazza E, Vink J, Bajka M, Wapner RJ, Hall TJ, House M. The mechanical role of the cervix in pregnancy. J Biomech 2015;48:1511–1523. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/J.JBIOMECH.2015.02.065&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 53. Obergasteiger J, Castonguay A-M, Pizzi S, Magnabosco S, Frapporti G, Lobbestael E, Baekelandt V, Hicks AA, Pramstaller PP, Gravel C, et al. The small GTPase Rit2 modulates LRRK2 kinase activity, is required for lysosomal function and protects against alpha-synuclein neuropathology. NPJ Parkinsons Dis 2023;9:44. 54. Oxvig C. The role of PAPP-A in the IGF system: location, location, location. J Cell Commun Signal 2015;9:177–187. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12079-015-0259-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25617049&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 55. Ozasa H, Tominaga T, Nishimura T, Takeda T. Lysyl oxidase activity in the mouse uterine cervix is physiologically regulated by estrogen. Endocrinology 1981;109:618–621. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1210/endo-109-2-618&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=6113953&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1981LZ52800040&link_type=ISI) 56. Papa A, Zakharov SI, Katchman AN, Kushner JS, Chen B-X, Yang L, Liu G, Jimenez AS, Eisert RJ, Bradshaw GA, et al. Rad regulation of CaV1.2 channels controls cardiac fight-or-flight response. Nat Cardiovasc Res 2022;1:1022–1038. 57. Parrettini S, Caroli A, Torlone E. Nutrition and Metabolic Adaptations in Physiological and Complicated Pregnancy: Focus on Obesity and Gestational Diabetes. Front Endocrinol 2020;11:611929. 58. Parry LJ, Vodstrcil LA. Relaxin physiology in the female reproductive tract during pregnancy. Adv Exp Med Biol 2007;612:34–48. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/978-0-387-74672-2_4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18161480&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251961000005&link_type=ISI) 59. Plomin R, DeFries JC, Knopik VS, Neiderhiser JM. Behavioral Genetics. 2012; Worth Publishers. 60. Polderman TJC, Benyamin B, Leeuw CA de, Sullivan PF, Bochoven A van, Visscher PM, Posthuma D. Meta-analysis of the heritability of human traits based on fifty years of twin studies. Nat Genet 2015;47:702–709. 61. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, Maller J, Sklar P, Bakker PIW de, Daly MJ, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 2007;81:559–575. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/519795&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17701901&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 62. Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science 2014;345:760–765. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNDUvNjE5OC83NjAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wNS8yNC8yMDI0LjA1LjIzLjI0MzA3ODA4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 63. Sheng YH, Hasnain SZ. Mucus and Mucins: The Underappreciated Host Defence System. Front Cell Infect Microbiol 2022;12:856962. 64. Sizova O, John LS, Ma Q, Molldrem JJ. Multi-faceted role of LRP1 in the immune system. Front Immunol 2023;14:1166189. 65. Smith GCS, Stenhouse EJ, Crossley JA, Aitken DA, Cameron AD, Connor JM. Early-pregnancy origins of low birth weight. Nature 2002;417:916. 66. Socha MW, Flis W, Pietrus M, Wartęga M, Stankiewicz M. Signaling Pathways Regulating Human Cervical Ripening in Preterm and Term Delivery. Cells [Internet] 2022;11.: 67. Solé-Navais P, Flatley C, Steinthorsdottir V, Vaudel M, Juodakis J, Chen J, Laisk T, LaBella AL, Westergaard D, Bacelis J, et al. Genetic effects on the timing of parturition and links to fetal birth weight. Nat Genet 2023;55:559–567. nature.com. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-023-01343-9&link_type=DOI) 68. Solovieff N, Cotsapas C, Lee PH, Purcell SM, Smoller JW. Pleiotropy in complex traits: challenges and strategies. Nat Rev Genet 2013;14:483–495. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrg3461&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23752797&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 69. Strauss JF, Romero R, Gomez-Lopez N, Haymond-Thornburg H, Modi BP, Teves ME, Pearson LN, York TP, Schenkein HA. Spontaneous preterm birth: advances toward the discovery of genetic predisposition. Am J Obstet Gynecol 2018;218:294–314.e2. Elsevier. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajog.2017.12.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 70. Svensson AC, Sandin S, Cnattingius S, Reilly M, Pawitan Y, Hultman CM, Lichtenstein P. Maternal effects for preterm birth: a genetic epidemiologic study of 630,000 families. Am J Epidemiol 2009;170:1365–1372. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/aje/kwp328&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19854802&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000272069900005&link_type=ISI) 71. Tait Wojno ED, Hunter CA, Stumhofer JS. The Immunobiology of the Interleukin-12 Family: Room for Discovery. Immunity 2019;50:851–870. 72. Tebar F, Chavero A, Agell N, Lu A, Rentero C, Enrich C, Grewal T. Pleiotropic Roles of Calmodulin in the Regulation of KRas and Rac1 GTPases: Functional Diversity in Health and Disease. Int J Mol Sci [Internet] 2020;21.: 73. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol 2003;3:133–146. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nri1001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12563297&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000181944800014&link_type=ISI) 74. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al. Proteomics. Tissue-based map of the human proteome. Science 2015;347:1260419. 75. Vega S de, Iwamoto T, Yamada Y. Fibulins: multiple roles in matrix structures and tissue functions. Cell Mol Life Sci 2009;66:1890–1902. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00018-009-8632-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19189051&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000268004300010&link_type=ISI) 76. Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, Yang J. 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet 2017;101:5–22. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2017.06.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28686856&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 77. Volozonoka L, Rots D, Kempa I, Kornete A, Rezeberga D, Gailite L, Miskova A. Genetic landscape of preterm birth due to cervical insufficiency: Comprehensive gene analysis and patient next-generation sequencing data interpretation. PLoS One 2020;15:e0230771. [journals.plos.org](http://journals.plos.org). 78. Wadon M, Modi N, Wong HS, Thapar A, O’Donovan MC. Recent advances in the genetics of preterm birth. Ann Hum Genet 2020;84:205–213. 79. Watanabe H, Minamino T. Genetics of Brugada syndrome. J Hum Genet 2016;61:57–60. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/jhg.2015.97&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26223181&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 80. Watanabe K, Taskesen E, Bochoven A van, Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat Commun 2017;8:1826. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-017-01261-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 81. Wolf HM, Latendresse SJ, Strauss JF III, Tarca AL, Romero R, Hassan SS, Webb BT, York TP. Risk factors for spontaneous preterm birth are mediated through changes in cervical length. medRxiv [Internet] 2023;Available from: [https://www.medrxiv.org/content/10.1101/2023.04.20.23288082.abstract](https://www.medrxiv.org/content/10.1101/2023.04.20.23288082.abstract). 82. Wolf HM, Romero R, Strauss JF, Hassan SS, Latendresse SJ, Webb BT, Tarca AL, Gomez-Lopez N, Hsu C-D, York TP. Study protocol to quantify the genetic architecture of sonographic cervical length and its relationship to spontaneous preterm birth. BMJ Open 2022;12:e053631. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYm1qb3BlbiI7czo1OiJyZXNpZCI7czoxMjoiMTIvMy9lMDUzNjMxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDUvMjQvMjAyNC4wNS4yMy4yNDMwNzgwOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 83. Wray S, Prendergast C, Arrowsmith S. Calcium-Activated Chloride Channels in Myometrial and Vascular Smooth Muscle. Front Physiol 2021;12:751008. 84. Wu T, Liu Z, Mak TSH, Sham PC. Polygenic power calculator: Statistical power and polygenic prediction accuracy of genome-wide association studies of complex traits. Front Genet 2022;13:989639. 85. Yamaoka K, Masuda K, Ogawa H, Takagi K, Umemoto N, Yasuoka S. Cloning and characterization of the cDNA for human airway trypsin-like protease. J Biol Chem 1998;273:11895–11901. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyNzMvMTkvMTE4OTUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wNS8yNC8yMDI0LjA1LjIzLjI0MzA3ODA4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 86. Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet 2011;88:76–82. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2010.11.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21167468&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 87. Yellon SM. Immunobiology of Cervix Ripening. Front Immunol 2019;10:3156. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2019.03156&link_type=DOI) 88. Yengo L, Vedantam S, Marouli E, Sidorenko J, Bartell E, Sakaue S, Graff M, Eliasen AU, Jiang Y, Raghavan S, et al. A saturated map of common genetic variants associated with human height. Nature 2022;610:704–712. 89. Yip C, Foidart P, Noël A, Sounni NE. MT4-MMP: The GPI-Anchored Membrane-Type Matrix Metalloprotease with Multiple Functions in Diseases. Int J Mol Sci [Internet] 2019;20.: 90. Yockey LJ, Iwasaki A. Interferons and Proinflammatory Cytokines in Pregnancy and Fetal Development. Immunity 2018;49:397–412. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.immuni.2018.07.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30231982&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 91. York TP, Eaves LJ, Lichtenstein P, Neale MC, Svensson A, Latendresse S, Långström N, Strauss JF. Fetal and maternal genes’ influence on gestational age in a quantitative genetic analysis of 244,000 Swedish births. Am J Epidemiol 2013;178:543–550. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/aje/kwt005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23568591&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000323045400005&link_type=ISI) 92. York TP, Eaves LJ, Neale MC, Strauss JF III. The contribution of genetic and environmental factors to the duration of pregnancy. Am J Obstet Gynecol 2014;210:398–405. Elsevier Inc. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajog.2013.10.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24096276&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 93. York TP, Strauss JF, Neale MC, Eaves LJ. Racial differences in genetic and environmental risk to preterm birth. PLoS One 2010;5:e12391. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0012391&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20811627&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 94. Zhang G, Feenstra B, Bacelis J, Liu X, Muglia LM, Juodakis J, Miller DE, Litterman N, Jiang P-P, Russell L, et al. Genetic Associations with Gestational Duration and Spontaneous Preterm Birth. N Engl J Med 2017;377:1156–1167. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1612665&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28877031&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F05%2F24%2F2024.05.23.24307808.atom) 95. Zhou P, Wang J, Wang J, Liu X. When autophagy meets placenta development and pregnancy complications. Front Cell Dev Biol 2024;12:1327167.