Next Article in Journal
Hypoxia-Inducible Factor in Renal Cell Carcinoma: From Molecular Insights to Targeted Therapies
Previous Article in Journal
Maximal Fat Oxidation During Exercise in Healthy Individuals: Lack of Genetic Association with the FTO rs9939609 Polymorphism
Previous Article in Special Issue
Loss of ZC4H2, an Arthrogryposis Multiplex Congenita Associated Gene, Promotes Osteoclastogenesis in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Etiology in Pelvic Organ Prolapse: Role of Connective Tissue Homeostasis, Hormone Metabolism, and Oxidative Stress

by
Wenxuan Jiang
1,
Rachel Yau Kar Cheung
1,
Cheuk Yan Chung
1,
Symphorosa Shing Chee Chan
1 and
Kwong Wai Choy
1,2,*
1
Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
2
Baylor College of Medicine Joint Center for Medical Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China
*
Author to whom correspondence should be addressed.
Submission received: 13 November 2024 / Revised: 15 December 2024 / Accepted: 22 December 2024 / Published: 24 December 2024
(This article belongs to the Special Issue Genetic Advances and Challenges in Complex Diseases)

Abstract

:
Background: Pelvic organ prolapse (POP) has become a common health problem among the aging population and affects an increasing number of elderly women worldwide. Studies within family and twin pairs provided strong evidence for the contribution of genetic factors to POP. Given the incomplete penetrance, polygenic traits, and small effect sizes of each variant in complex diseases, it is not always easy to evaluate the genetic susceptibility and molecular mechanisms involved in POP. Objectives: This review intends to comprehensively summarize the current studies on genetic variants associated with POP. Methods: We performed a comprehensive review to summarize the genetic findings from genome-linkage studies, genome-wide association studies, candidate association studies, and gene expression analyses. Results: We summarized genetic variants associated with connective tissue homeostasis, hormone metabolism, and oxidative stress, which were potentially related to the pathophysiology of POP. We also reviewed the limited polygenic risk score (PRS) studies generated for each individual’s genetic risk stratification and its integration into clinical risk factors for disease prediction. Conclusions: This pooled analysis provides moderate epidemiological credibility for associations of these genetic variants with POP to bridge the gap between genetic research and clinical medicine towards understanding the genetic etiology of POP. It also highlights the potential of PRS as a risk prediction model.

1. Introduction

Pelvic organ prolapse (POP) is the descent of one or more pelvic organs due to the weakness or loss of pelvic floor support from connective tissues, muscles, or both [1]. It has attracted much attention given its high prevalence, significant impairment of quality of life, and heavy emotional burden in elderly women [2,3,4]. In the United States, about 3% of women were affected by symptomatic POP [5]. In China, the prevalence of symptomatic POP was 9.6%, according to a nationwide epidemiological survey [6]. Aging in the population is a global issue that can exacerbate the burden of the economy and public healthcare systems [7]. The cumulative risk of POP surgeries was 12.6% by the age of 80 years, and the annual risk of POP surgeries increased with age until a peak of 4.3 per 1000 women between 71 and 73 years old [8]. It was predicted that, by 2050, the number of surgeries for POP will increase significantly with the aging population [9]. Therefore, understanding the pathophysiology of POP is important for us to identify women at increased risk and provide prevention and intervention strategies. Age-related biological deterioration can occur in different types of tissues and has been recognized as the main risk factor for human diseases, which results from the accumulation of cellular and molecular damage [10].
Human aging is a complex process affected by genetic and environmental factors [11]. There is a normal decline in pelvic floor function with aging. During this period, the risk factors related to the weakening or injury of pelvic floor connective tissues or muscles will accelerate the deterioration of function. The well-known factors include pregnancy, vaginal childbirth, frequency of childbirth, advanced age, hormone changes, ethnicity, increased intra-abdominal pressure, smoking, family history, and previous pelvic surgery [12]. These factors usually contribute to POP by interacting with each other rather than alone. To better understand the role of these risk factors, a “lifespan model” [13] integrated the risk factors involved physiology, anatomy, genetics, and lifestyles into three major human life phases to describe how these factors contribute to POP at different life phases and what independent, interactive, and cumulative effects they have on pelvic floor function. During an individual’s growth and development of pelvic floor function, predisposing factors could make an individual susceptible to the disease. An individual with good functional reserve may not develop POP during her lifespan, while an individual with less functional reserve may suffer from POP late or early in life based on age-related normal decline. During pregnancy and childbirth, under different predisposing conditions or interventions, some small pelvic floor injuries may completely recover, while some significant or severe pelvic floor injuries can only partially recover or cannot recover. Genetic factors can act as intrinsic drivers that regulate the growth of pelvic floor and respond to the dramatic and dynamic changes. During the aging phase, genetically programmed variations in aging, hormonal decline, and lifestyle (such as smoking and chronic cough) can cause variations in the rate of the pelvic floor function decline. Therefore, it is highlighted that genetic and environmental factors should be considered when analyzing POP causation.
In this review, we focus on the genetic factors involved in the development of POP. Recently, the International Urogynecology Consultation (IUC) evaluated the impact of genetics on POP across different studies and suggested that genetic factors had a strong etiologic link to POP [14]. Current studies are devoted to identifying the predisposed genetic variants associated with POP. Given the incomplete penetrance, polygenic traits, and small effect sizes of each variant in complex diseases, the evaluation of genetic susceptibility and molecular mechanisms involved in POP is not easy [15]. Therefore, this review intends to comprehensively summarize the current studies on genetic variants associated with POP combining with the findings from genome linkage studies, genome-wide association studies (GWASs), and candidate gene association studies in three different aspects: genetic variants associated with connective tissue homeostasis, hormone metabolism, and oxidative stress (OS).

2. A Familial or Genetic Basis of POP

Current epidemiological evidence demonstrates the heritability of POP, and the results are consistent between several studies. Family studies showed that prolapse could be inherited in an autosomal dominant fashion with incomplete penetrance [16]. There was also a high concordance (74.3% to 91.1%) in POP stage between nulliparous women and their parous sisters [17]. Two large, population-based studies found the relative risk (RR) of POP increased in first- and third-degree female relatives (4.15 and 1.24, respectively) and elevated from 2.36 to 6.26 with the increased number of affected first-degree relatives [18,19]. A large twin study showed that genetic, non-shared environmental, and shared environmental factors contributed to around 40%, 40%, and 20% of the variance of POP, respectively [20]. Several systematic reviews and meta-analyses showed that a positive family history of POP was associated with a 2.3- to 2.7-fold increased risk for POP as well as a 1.4- to 1.8-fold increased risk for POP recurrence [21,22,23]. These studies revealed a familial or genetic basis for POP.

3. Genetic Variants Associated with Remodeling of Extracellular Matrix (ECM) in Pelvic Floor Connective Tissues

ECM is a series of macromolecules composed mainly of proteins and polysaccharides, including collagens, elastin, non-collagenous glycoproteins, and proteoglycans/glycosaminoglycans [24]. ECM proteins form complex matrix structures (lamina, collagen, and elastin fibers) through direct interactions or lysyl oxidase-mediated cross-linking to maintain the strength, which is essential for maintaining homeostasis [25,26]. In addition, the degradation of extracellular proteases or the cleavage of matrix proteins can contribute to ECM degradation and remodeling [27,28]. The balance of anabolism and catabolism can keep the homeostasis of pelvic floor connective tissues and provide support for pelvic organs. The significant findings are listed in Table 1.
Collagen Collagen is the main component (70–80%) of connective tissues [45]. Type I collagen and type III collagen are the main components. Type I collagen is responsible for mechanical strength, while type III collagen is responsible for tissue elasticity and extensibility [46]. Type IV collagen is the network-forming collagen that constitutes the sheet-like structure with other components [47]. Type XVIII collagen plays a role in the structure composition of the basement membranes to strengthen its structure and provide additional anchor sites [48]. Only rs1800255 of COL3A1 was significantly associated with POP in two independent cohorts, and it was also supported by a system review and meta-analysis [29,30,49,50]. The substitution of an adenine (A) to guanidine (G) in position 2092 (exon 30) causes a missense functional variant, which leads to the disturbance of triple helical conformation of type III collagen [30]. The rs2236479 of COL18A1 was firstly reported in a genome-wide association study by Allen-Brady et al. [44]. However, this association was not replicated in another two independent cohorts including Brazilian, African American, and Hispanic [51,52] (Supplementary Table S1). Several single nucleotide polymorphisms (SNPs) of COL4A2, COL5A1, and COL14A1 were only identified in one small East Asian cohort [31].
Elastic fibers and lysyl oxidase Elastic fiber is another important component of the pelvic connective tissues, which is exposed to extraordinary forces and provides stretching, resilience, and cell interactivity to pelvic floor tissues. Elastin and microfibrils are the main components of elastic fibers, and elastin accounts for up to 90% of their mass [53]. Elastin synthesis is a complex process, which is formed by the hierarchical assembly of its monomer (tropoelastin) [54]. Tropoelastin is secreted by elastogenic cell types and assembles into consistent globules of several microns in diameter. After excretion into the extracellular space, it undergoes rapid ordered assembly and cross-linking into elastin and is deposited on the microfibril surface [55].
Fibulins play a role in their assembly and microfibrillar deposition [54], especially fibulin-5, without which assembly would not be possible. The fibulin-5 protein is encoded by the fibulin 5 (FBLN5) gene. The associations of rs12589592 and rs2018736 of FBLN5 with POP were validated in a Russian cohort and a Chinese minority population (Table 1 and Supplementary Table S1) [32,33]. Animal studies demonstrated that Fbln5 knockout mice had a dilated vaginal wall, enlarged genital hiatus, prolapse at more than 6 months of age, and increased severity of prolapse within 1 week postpartum [56]. In addition, fibulin-3, another component of fibulins, is encoded by the fibulin 3 gene (EFEMP1 gene, also known as FBLN3). Two genome-wide association studies also identified three SNPs related to the EFEMP1 gene, namely, rs1430191, rs11899888, and rs3791675 [41,42]. However, these associations were not replicated in other European and Japanese cohorts [43,57].
Lysyl oxidase (LOX) is also involved in this assembly process as a catalyst [58]. The abnormal synthesis of LOX or LOX-like (LOXL) proteins leads to an altered elastogenesis. LOXL proteins 1–4 are encoded by different clusters of LOXL genes. Two variants, rs4886778 of LOXL1 from a candidate gene association study and rs2862296 of LOXL4 from a genome-wide association study, showed associations with POP [34,42]. Moreover, LOXL4 gene, located on chromosome 10q24.2, was one of the candidate genes in the chromosome 10q24-26 region. This region was identified in a genetic linkage analysis (heterogeneity logarithm of odds [HLOD] score = 3.40) [59]. HLOD is a statistical estimate of the probability that two nearby loci are likely to be inherited together. An HLOD score higher than 3.0 is generally accepted as evidence for genetic linkage [60]. An animal study also supported that Loxl1-deficient mice underwent pelvic prolapse within 1–2 days postpartum and prominent pelvic descent remained over time [61].
Laminin Laminin is a type of glycoproteins that forms the major non-collagenous portion of the basement membranes. Its isoforms are composed of three nonidentical chains: α, β, and γ [62]. Most isoforms contain the laminin subunit γ-1, which is encoded by LAMC1 gene. The first genome-wide linkage analysis reported that rs10911193, located at the promoter of LAMC1, was associated with early-onset prolapse, potentially by affecting the binding site of the transcription factor NFIL3 (nuclear factor, interleukin 3 regulated) [63]. However, further analysis could not identify this association in different populations [33,35,64,65]. In addition, rs10911241 of LAMC1 was shown to associate with POP in a Chinese cohort [35].
Extracellular proteases Many proteases exist in the extracellular space and directly interact with the ECM, causing ECM degradation and remodeling. Degradation not only decreases the quantity of matrix proteins but also produces matrix protein degradation-derived bioactive fragments [27]. The main proteases that mediate ECM degradation include MMPs (matrix metalloproteinases), ADAMs (disintegrin and metalloproteinases), and ADAMTSs (disintegrin and metalloproteinases with thrombospondin motifs). Collagen, elastin, laminins, and proteoglycans can be degraded by these proteases as substrates. Another type of proteases, TIMPs (tissue inhibitors of matrix metalloproteinases), can inhibit the activities of MMPs, ADAMs, and ADAMTSs [66]. Several SNPs of MMP1, MMP9, and MMP10 were associated with POP in candidate gene association studies [36,37,38,39]. Genome-wide association studies also identified the associations of rs42400 of ADAMTS16, rs10810888 of ADAMTSL1, rs235929 near ADAMTS5, and ADAMTS1 with POP [42], which overlapped with hernia-associated genetic loci [67]. Other SNPs of ADAMTS1, ADAMTS13, and TIMP2 were only found to have associations with POP in a small Chinese cohort [40]. In addition, the TIMP2 gene, located on chromosome 17q25.3, was one of the candidate genes in the chromosome 17q25 region. This region was identified in a genetic linkage analysis (HLOD score = 3.30) [59].
In addition to the above genetic variants, some novel variants possibly related to ECM homeostasis were also identified in genome-wide association studies [42]. The rs9306894 was located at the 3’UTR region of GDF7 gene (also known as BMP12 gene). This gene might play a role in the differentiation of the tendon cells and promote the synthesis of type I and III collagen, which was associated with connective tissue homeostasis [68,69]. The rs11031796 of WT1-AS was also reported in hernia, which shared a similar pathogenesis with POP [70]. The other two loci related to the WT1 gene were also reported in European and East Asian cohorts [43]. HOXD13 around rs77648136 and HOXA11 were reported to be involved in the regulation of collagen by regulating the activity of MMPs [71,72]. These findings highlighted a genetic correlation with abnormal ECM remodeling, and this progressive remodeling would contribute to the POP.

4. Genetic Variants Associated with Hormone Metabolism

The decline in sex hormone levels with menopause in aging women has negative effects on the female urogenital system, leading to atrophic changes. The strength and flexibility of pelvic connective tissues and muscles can also be affected by sex hormones that interact with specific receptors [73]. The sex hormone receptor is a type of nuclear hormone receptor. It is involved in transcription modulation through the ligand-binding domain (LBD) and DNA-binding domain (DBD). These two highly conserved regions can transmit activated steroid receptors from the cytoplasm to the nucleus and act on transcription regulation [74]. Except for hormone receptors, other factors related to sex hormone metabolism can also have effects on pelvic floor connective tissues. The significant findings are shown in Table 2.
Estrogen receptors include estrogen receptor α (ER-α) and estrogen receptor β (ER-β), which are encoded by the ESR1 gene and ESR2 gene. Estrogen regulates the transcriptional signature of its target tissues via the nuclear pathway activated by binding to ER-α and ER-β [77]. The abundance of estrogen receptors in the urogenital tract may explain why the natural reduction in endogenous estrogen can cause or potentiate pelvic floor disorders (PFDs) [78]. Only rs17847075 (rs2077647), rs2234693, and rs2228480 of ESR1 were found to be associated with POP in Chinese and Ashkenazi-Jewish origin populations [33,65,75]. Among them, the association of rs2228480 with POP was also supported by a systematic review and meta-analysis [49]. There was no association identified between ESR2 and POP (Supplementary Table S1). Only two haplotypes of ESR2 were associated with POP [79]. For other types of hormone receptors, only the rs484389 located in the 3’-UTR region of progesterone receptor (PGR) was found to be significantly associated with POP in Chinese Taiwanese women [76].
In addition, rs3820282 of WNT4 was identified to be associated with POP in genome-wide association studies [41,42]. This variant was also reported to increase the risk of some estrogen-related diseases [80,81]. WNT4 is a transcription factor involved in the Wnt signaling pathway. It encodes the protein that participates in the regulation of female sexual differentiation and the development of the female reproductive tract [82]. The loss of function of WNT4 genes leads to a partial sex reversal in humans [83]. DVL2 (rs72839768 mapped gene) was also reported to be involved in the Wnt signaling pathway [84,85].

5. Genetic Variants Associated with Oxidative Stress That Disturbs Cellular Homeostasis in Pelvic Floor Support Tissues

Oxidative stress (OS) is caused by an imbalance in reactive oxygen species (ROS) and antioxidant defense systems in cells, tissues, or organs [86]. The source of ROS is mainly from the mitochondrial respiratory chain [87]. If their balance is destroyed, ROS can react spontaneously on biomolecules, such as DNA, RNA, protein, and lipids, leading to cell death and disease. Therefore, OS interferes with the process of collagen and elastin synthesis [88] (Table 3).
As shown in Table 3, two polymorphisms of OS-related genes, rs1695 of glutathione S-transferase pi 1 (GSTP1) and rs1136410 of poly (ADP-ribose) polymerase 1 (PARP1), showed significant associations with POP in the Korean population [89,90]. Glutathione S-transferases participate in the major detoxification mechanisms in humans by combining with a wide range of electrophilic compounds. PARP1 is involved in the base excision repair of oxidative DNA damage and acts as a mediator to direct cell fates, according to the type and strength of stress [89,91]. Another three associated variants, rs1810636 near IDH3B, rs1036819 of ZFAT, and rs2267372 of MAFF, were also identified in genome-wide association studies [42,44]. The protein encoded by IDH3B is the β subunit of one isozyme of NAD(+)-dependent isocitrate dehydrogenase. It can participate in energy production by mitochondria [85]. ZFAT encodes a protein that likely binds to DNA and functions as a transcriptional regulator involved in apoptosis and cell survival [85]. MAFF encodes a basic leucine zipper (bZIP) transcription factor and acts as the OS reactive protein in the inflammation process [85,92].

6. Other Genetic Findings Related to Predisposition of POP

Genome-wide association studies showed a positive correlation between metabolic and cardiovascular health and POP (rs4779517 of KLF13, rs12314243 of DUSP16, rs10762631 of VCL, rs7072877 near FGFR2, and rs73197353 and rs1247943 near TBX5) (Table 3) [41,42,43,92,93,94]. It indicated that the comorbidities could be risk factors of POP [95,96]. Several significant loci were identified, but the potential biological functions had not been demonstrated (Table 3). Other non-significant associations in different cohorts are listed in Supplementary Table S1 [29,30,31,32,33,34,35,36,37,38,39,40,51,63,64,65,75,76,79,88,97,98,99,100,101,102,103,104,105,106,107,108,109]. The distribution of all the significant genetic variants in chromosomes is summarized in Figure 1.

7. Gene Expression of POP-Associated Genes

Gene expression is an intermediate linkage between the DNA sequence and phenotype by RNA transcription and protein translation [110]. The human genome regulates these expression processes in target tissues, which varies across cell types and individuals [111]. Pelvic floor support structures mainly include ligaments, especially cardinal ligaments (CLs) and uterosacral ligaments (USLs), vaginal wall, levator ani muscle (LAM), and pelvic fasciae [112]. The increased or decreased gene expressions in these tissues may be related to the changes in pelvic floor functions. The expression of the above POP-associated genes in these pelvic floor tissues is shown in Table 4.
Corresponding to the above genetic findings, for ECM-related components, there were different results regarding the RNA expression of COL3A1 gene and the protein expression of type III collagen across different studies. The increased or decreased expressions were found in CLs, USLs, and the vaginal wall of POP subjects in different studies. However, there was no difference in round ligaments (RLs) and para-urethral tissues between POP subjects and controls [26,45,113,114,115,116,117,118,119,120,121,122,124,125,126,127,148]. The protein expression of elastin was decreased in CLs, USLs, and the vaginal wall of POP subjects in some studies [26,58,120,121,123,128,129]. The RNA expression of FBLN5 gene and the protein expression of fibulin-5 were significantly decreased in the different tissues of POP subjects [58,113,118,130,132,133,134,135,136]. In addition, there were no differences in the RNA expression of EFEMP1 and LOXL4 gene and protein expression on type IV collagen, type V collagen, fibulin-3, and laminin across different tissues between POP subjects and controls [58,119,120,130,131,132,134,136]. For extracellular proteases, most studies showed an increased RNA and protein expression of MMP1, MMP9, and MMP10 and a decreased RNA expression of TIMP2 across different tissues in POP subjects [113,116,119,121,125,137,138,139,140,141,142,143,144,145]. For hormone receptors, the RNA expression of ESR1 and protein expression of ER-α decreased in the USLs and RLs of POP cases [115,116,145]. However, one study found both of them increased in USLs in POP subjects [147]. The RNA and protein expressions of PGR were inconsistent in USLs [146,147]. The expressions of other genes and products of OS are shown in Supplementary Table S2 [26,45,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,136,137,138,141,142,143,144,145,147,148,149,150,151,152,153,154,155,156,157,158,159,160]. There was also an increased level of products of OS across different tissues in POP subjects. The significant SNPs and related gene expression in different pelvic floor tissues are shown in Figure 2. These results indicated the heterogeneity of gene expression in different tissues and cells across different populations.
Single-cell RNA sequencing (scRNA-seq) enables a deep understanding of cellular heterogeneity to some extent by analyzing all cell populations, cell-to-cell communication, and averaged gene expression at the single-cell level [161]. There were only limited studies on POP. The three most abundant cell types in the vaginal wall of POP women identified in current scRNA-seq studies were fibroblast (55.49%), smooth muscle cells (SMCs) (17.97%), and macrophages (7.51%). The genes or transcription factors in these cell types were involved in dysregulated ECM organization and immune reaction. The increased immune regulation and tissue remodeling-related interactions between these three cell types suggested that the phenotypic switch from SMCs to myofibroblasts could be the underlying cause of structural changes in the muscularis of the vaginal wall [162]. During this process, ECM organization and antigen presentation were also enhanced [163]. The age-related difference in the biological process in the vaginal wall revealed that the upregulated biological process in old POP women was mainly related to chronic inflammation, while the upregulated biological process in young POP women was mainly related to ECM metabolism [164]. Another scRNA-seq study identified that the three most abundant cell types of USLs in POP women were SMCs (34.93%), endothelial cells (21.29%), and fibroblasts (17.36%) [165]. A significant reduction in receptor–ligand pairs between immune cells and fibroblasts and cell adhesion between fibroblasts and endothelial cells were also demonstrated in the USLs of POP women.

8. Polygenic Risk Score (PRS) and Interaction

Although a large number of genetic variants (mostly SNPs) were identified in different association studies of POP, each variant typically has a small effect and low predictive value. A polygenic risk score (PRS) can aggregate the effects of all variants across the genome and quantify the individual’s genetic risk of developing a certain disease [15]. It is calculated by the sum of multiple risk alleles, each weighted by the corresponding effect size derived from GWAS [166]. Based on the findings from this largest genome-wide association study, the first PRS of POP was generated for each individual’s genetic risk stratification. The PRS model showed a similar prediction value with clinical joint models (five established clinical factors combined: number of children, body mass index, ever smoked, constipation, and asthma) (concordance index [Harrell C-statistic]: 0.583 ± 0.007 vs. 0.588 ± 0.007). The poor concordance index of PRS might be due to the missing heritability, such as undiscovered SNP heritability, rare variants or structural variants that are not included in the PRS, gene–gene interactions, age-dependent effects, phenotype definition, and heterogeneity of the condition, while the lower concordance index of clinical joint models might be limited by the unavailability of other clinical risk factors that would improve the predictive ability in the database, such as newborn information and mode of delivery. In addition, adding PRS to the clinical joint model could improve the prediction of the combined model (Harrell C-statistic: 0.630 ± 0.007, +4.2 percentage points) [42]. This finding illustrated PRS as a tool to evaluate individual risk prediction, but further studies need to consider adding more available factors into PRS and the clinical joint model to advance the concept of using genomic information to stratify disease risk in gynecological conditions.

9. Challenges and Prospects

Although current studies have identified many genetic variants associated with POP, and some related genes are expressed in pelvic floor tissues, the discordant results in different studies are still the major questions to evaluate its genetic risk. For candidate gene association studies, the inconsistencies may be explained by the small sample size and different ethnic populations. For genome-wide association studies, the “missing heritability” may be explained by common variants with small effects, rare variants with large effect sizes that are not included in the analyses, and a combination of genotypic, environmental, and epigenetic interactions [167,168]. Moreover, the identified SNPs only account for a small fraction of the genetic component, which cannot fully explain the picture of genetic characteristics [169]. Copy number variants (CNVs) are deletions or duplications of genomic fragments of more than 50 base pairs. They cover around 4.8–9.5% of the human genome and encompass more nucleotide variations than SNPs [170]. Therefore, it is proposed that CNVs can at least explain part of the “missing heritability”. Although genome-wide CNV association analyses have been applied in certain common complex diseases and successfully identified CNVs related to the risk of diseases, they are relatively limited in obstetric and gynecological diseases, especially in pelvic floor disorders. In the future, CNV-associated studies can be performed in different populations.
Current studies cannot fully correlate the genetic findings with the corresponding gene expression in pelvic floor tissues. Further studies of genetic variants in different tissues are still needed to validate these findings. Apart from the direct alteration of the DNA sequence, gene expression can also be affected by the heritable modification without any change in the DNA sequence, which is called epigenetics, including DNA methylation, histone regulation, and non-coding RNA [171]. Although current epigenetic studies on POP have identified the association of DNA methylation patterns and microRNA with POP [172,173,174], the impact of these modifiers in the development of POP and the components of pelvic floor tissues has not been demonstrated. It is necessary to consider these effects on gene expression in future studies. In addition, further studies on the relationship between genetic factors involved in connective tissues, hormone metabolism, oxidative stress and environmental factors can be performed for better understanding the contributions of genetic and environmental factors to POP.

10. Conclusions

Based on the current evidence, it is believed that genetic factors have a strong etiologic linkage to POP. Single nucleotide polymorphisms related to extracellular matrix homeostasis, hormone metabolism, and oxidative stress are associated with the risk of POP. The implementation of combined models with genetic and clinical risk factors can be used for each individual’s POP risk stratification. However, validation in a larger cohort is needed. Gene expression studies and epigenetic studies are also necessary for understanding their roles in the maintenance of normal functions across different tissues. Further genome-wide association studies involving copy number variants should be performed to resolve the “missing heritability” in POP.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes16010005/s1, Table S1: Summary of non-significant genetic loci in candidate gene association studies; Table S2: Summary of gene expression of other genes in different tissues; Table S3: Gene transcript.

Author Contributions

W.J. and K.W.C. wrote the original version of the manuscript; R.Y.K.C., C.Y.C. and S.S.C.C. reviewed and edited the original version of the manuscript; W.J. and K.W.C. edited the final versions of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received support from Liauw’s Family Reproductive Genomics Fund.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Barber, M.D. Pelvic organ prolapse. BMJ 2016, 354, i3853. [Google Scholar] [CrossRef] [PubMed]
  2. Ghetti, C.; Skoczylas, L.C.; Oliphant, S.S.; Nikolajski, C.; Lowder, J.L. The emotional burden of pelvic organ prolapse in women seeking treatment: A qualitative study. Urogynecology 2015, 21, 332–338. [Google Scholar] [CrossRef]
  3. Sanses, T.V.; Schiltz, N.K.; Richter, H.E.; Koroukian, S.M. Trends and factors influencing inpatient prolapse surgical costs and length of stay in the United States. Urogynecology 2016, 22, 103–110. [Google Scholar] [CrossRef]
  4. Robinson, D.; Prodigalidad, L.T.; Chan, S.; Serati, M.; Lozo, S.; Lowder, J.; Ghetti, C.; Hullfish, K.; Hagen, S.; Dumoulin, C. International Urogynaecology Consultation chapter 1 committee 4: Patients’ perception of disease burden of pelvic organ prolapse. Int. Urogynecol. J. 2022, 33, 189–210. [Google Scholar] [CrossRef]
  5. Wu, J.M.; Vaughan, C.P.; Goode, P.S.; Redden, D.T.; Burgio, K.L.; Richter, H.E.; Markland, A.D. Prevalence and trends of symptomatic pelvic floor disorders in US women. Obstet. Gynecol. 2014, 123, 141–148. [Google Scholar] [CrossRef]
  6. Pang, H.; Zhang, L.; Han, S.; Li, Z.; Gong, J.; Liu, Q.; Liu, X.; Wang, J.; Xia, Z.; Lang, J. A nationwide population-based survey on the prevalence and risk factors of symptomatic pelvic organ prolapse in adult women in China–a pelvic organ prolapse quantification system-based study. BJOG Int. J. Obstet. Gynaecol. 2021, 128, 1313–1323. [Google Scholar] [CrossRef]
  7. Fang, E.F.; Scheibye-Knudsen, M.; Jahn, H.J.; Li, J.; Ling, L.; Guo, H.; Zhu, X.; Preedy, V.; Lu, H.; Bohr, V.A. A research agenda for aging in China in the 21st century. Ageing Res. Rev. 2015, 24, 197–205. [Google Scholar] [CrossRef] [PubMed]
  8. Wu, J.M.; Matthews, C.A.; Conover, M.M.; Pate, V.; Funk, M.J. Lifetime risk of stress urinary incontinence or pelvic organ prolapse surgery. Obstet. Gynecol. 2014, 123, 1201–1206. [Google Scholar] [CrossRef] [PubMed]
  9. Wu, J.M.; Kawasaki, A.; Hundley, A.F.; Dieter, A.A.; Myers, E.R.; Sung, V.W. Predicting the number of women who will undergo incontinence and prolapse surgery, 2010 to 2050. Am. J. Obstet. Gynecol. 2011, 205, 230.e231–230.e235. [Google Scholar] [CrossRef]
  10. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef]
  11. May, M. Biomarkers of aging remain elusive as researchers try to slow the biological clock. Nat. Med. 2023, 29, 2673–2676. [Google Scholar] [CrossRef]
  12. Jelovsek, J.E.; Maher, C.; Barber, M.D. Pelvic organ prolapse. Lancet 2007, 369, 1027–1038. [Google Scholar] [CrossRef] [PubMed]
  13. DeLancey, J.O.; Low, L.K.; Miller, J.M.; Patel, D.A.; Tumbarello, J.A. Graphic integration of causal factors of pelvic floor disorders: An integrated life span model. Am. J. Obstet. Gynecol. 2008, 199, 610.e611–610.e615. [Google Scholar] [CrossRef]
  14. Deprest, J.A.; Cartwright, R.; Dietz, H.P.; Brito, L.G.O.; Koch, M.; Allen-Brady, K.; Manonai, J.; Weintraub, A.Y.; Chua, J.W.; Cuffolo, R. International Urogynecological Consultation (IUC): Pathophysiology of pelvic organ prolapse (POP). Int. Urogynecol. J. 2022, 33, 1699–1710. [Google Scholar] [CrossRef] [PubMed]
  15. Purcell, S.M.; Wray, N.R.; Stone, J.L.; Visscher, P.M.; O’Donovan, M.C.; Sullivan, P.F.; Sklar, P. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature 2009, 460, 748–752. [Google Scholar] [CrossRef]
  16. Jack, G.S.; Nikolova, G.; Vilain, E.; Raz, S.; Rodríguez, L.V. Familial tranmission of genitovaginal prolapse. Int. Urogynecol. J. 2006, 17, 498–501. [Google Scholar] [CrossRef] [PubMed]
  17. Buchsbaum, G.M.; Duecy, E.E.; Kerr, L.A.; Huang, L.-S.; Perevich, M.; Guzick, D.S. Pelvic organ prolapse in nulliparous women and their parous sisters. Obstet. Gynecol. 2006, 108, 1388–1393. [Google Scholar] [CrossRef] [PubMed]
  18. Norton, P.A.; Allen-Brady, K.; Cannon-Albright, L.A. The familiality of pelvic organ prolapse in the Utah Population Database. Int. Urogynecol. J. 2013, 24, 413–418. [Google Scholar] [CrossRef] [PubMed]
  19. Allen-Brady, K.; Norton, P.A.; Hill, A.J.; Rowe, K.; Cannon-Albright, L.A. Risk of pelvic organ prolapse treatment based on extended family history. Am. J. Obstet. Gynecol. 2020, 223, 105.e101–105.e108. [Google Scholar] [CrossRef] [PubMed]
  20. Altman, D.; Forsman, M.; Falconer, C.; Lichtenstein, P. Genetic influence on stress urinary incontinence and pelvic organ prolapse. Eur. Urol. 2008, 54, 918–923. [Google Scholar] [CrossRef] [PubMed]
  21. Lince, S.L.; van Kempen, L.C.; Vierhout, M.E.; Kluivers, K.B. A systematic review of clinical studies on hereditary factors in pelvic organ prolapse. Int. Urogynecol. J. 2012, 23, 1327–1336. [Google Scholar] [CrossRef]
  22. Friedman, T.; Eslick, G.D.; Dietz, H.P. Risk factors for prolapse recurrence: Systematic review and meta-analysis. Int. Urogynecol. J. 2018, 29, 13–21. [Google Scholar] [CrossRef]
  23. Samimi, P.; Jones, S.H.; Giri, A. Family history and pelvic organ prolapse: A systematic review and meta-analysis. Int. Urogynecol. J. 2021, 32, 759–774. [Google Scholar] [CrossRef] [PubMed]
  24. Fu, Y.; Zhou, Y.; Wang, K.; Li, Z.; Kong, W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ. Res. 2024, 134, 931–949. [Google Scholar] [CrossRef] [PubMed]
  25. del Monte-Nieto, G.; Fischer, J.W.; Gorski, D.J.; Harvey, R.P.; Kovacic, J.C. Basic biology of extracellular matrix in the cardiovascular system, part 1/4: JACC focus seminar. J. Am. Coll. Cardiol. 2020, 75, 2169–2188. [Google Scholar] [CrossRef] [PubMed]
  26. Ewies, A.A.; Al-Azzawi, F.; Thompson, J. Changes in extracellular matrix proteins in the cardinal ligaments of post-menopausal women with or without prolapse: A computerized immunohistomorphometric analysis. Hum. Reprod. 2003, 18, 2189–2195. [Google Scholar] [CrossRef]
  27. Ricard-Blum, S.; Vallet, S.D. Fragments generated upon extracellular matrix remodeling: Biological regulators and potential drugs. Matrix Biol. 2019, 75, 170–189. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, B.; Yeh, J. Alterations in connective tissue metabolism in stress incontinence and prolapse. J. Urol. 2011, 186, 1768–1772. [Google Scholar] [CrossRef] [PubMed]
  29. Chen, H.-Y.; Chung, Y.-W.; Lin, W.-Y.; Wang, J.-C.; Tsai, F.-J.; Tsai, C.-H. Collagen type 3 α 1 polymorphism and risk of pelvic organ prolapse. Int. J. Gynecol. Obstet. 2008, 103, 55–58. [Google Scholar] [CrossRef]
  30. Kluivers, K.B.; Dijkstra, J.R.; Hendriks, J.C.; Lince, S.L.; Vierhout, M.E.; van Kempen, L.C. COL3A1 2209G> A is a predictor of pelvic organ prolapse. Int. Urogynecol. J. 2009, 20, 1113–1118. [Google Scholar] [CrossRef] [PubMed]
  31. Li, L.; Sun, Z.; Chen, J.; Zhang, Y.; Shi, H.; Zhu, L. Genetic polymorphisms in collagen-related genes are associated with pelvic organ prolapse. Menopause 2020, 27, 223–229. [Google Scholar] [CrossRef]
  32. Khadzhieva, M.B.; Kamoeva, S.V.; Chumachenko, A.G.; Ivanova, A.V.; Volodin, I.V.; Vladimirov, I.S.; Abilev, S.K.; Salnikova, L.E. Fibulin-5 (FBLN5) gene polymorphism is associated with pelvic organ prolapse. Maturitas 2014, 78, 287–292. [Google Scholar] [CrossRef]
  33. Abulaizi, A.; Abula, A.; Ababaikeli, G.; Wan, X.; Du, R.; Zhakeer, A. Identification of pelvic organ prolapse risk susceptibility gene SNP locus in Xinjiang women. Int. Urogynecol. J. 2020, 31, 123–130. [Google Scholar] [CrossRef]
  34. Ashikari, A.; Suda, T.; Miyazato, M. Collagen type 1A1, type 3A1, and LOXL1/4 polymorphisms as risk factors of pelvic organ prolapse. BMC Res. Notes 2021, 14, 15. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, J.; Li, L.; Lang, J.; Zhu, L. Common variants in LAMC1 confer risk for pelvic organ prolapse in Chinese population. Hereditas 2020, 157, 26. [Google Scholar] [CrossRef] [PubMed]
  36. Ferrari, M.M.; Rossi, G.; Biondi, M.L.; Vigano, P.; Dell’Utri, C.; Meschia, M. Type I collagen and matrix metalloproteinase 1, 3 and 9 gene polymorphisms in the predisposition to pelvic organ prolapse. Arch. Gynecol. Obstet. 2012, 285, 1581–1586. [Google Scholar] [CrossRef] [PubMed]
  37. Chen HueyYi, C.H.; Lin WeiYong, L.W.; Chen YungHsiang, C.Y.; Chen WenChi, C.W.; Tsai FuuJen, T.F.; Tsai ChangHai, T.C. Matrix metalloproteinase-9 polymorphism and risk of pelvic organ prolapse in Taiwanese women. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010, 149, 222–224. [Google Scholar] [CrossRef] [PubMed]
  38. Wu, J.M.; Visco, A.G.; Grass, E.A.; Craig, D.M.; Fulton, R.G.; Haynes, C.; Weidner, A.C.; Shah, S.H. Matrix metalloproteinase-9 genetic polymorphisms and the risk for advanced pelvic organ prolapse. Obstet. Gynecol. 2012, 120, 587–593. [Google Scholar] [CrossRef]
  39. Wang, H.; Zhang, Z.Q.; Wang, S.Z.; Lu, J.L.; Wang, X.L.; Zhang, Z.Y. Association of matrix metalloproteinase-10 polymorphisms with susceptibility to pelvic organ prolapse. J. Obstet. Gynaecol. Res. 2015, 41, 1972–1981. [Google Scholar] [CrossRef] [PubMed]
  40. Li, L.; Ma, Y.; Yang, H.; Sun, Z.; Chen, J.; Zhu, L. The polymorphisms of extracellular matrix-remodeling genes are associated with pelvic organ prolapse. Int. Urogynecol. J. 2022, 33, 267–274. [Google Scholar] [CrossRef]
  41. Olafsdottir, T.; Thorleifsson, G.; Sulem, P.; Stefansson, O.A.; Medek, H.; Olafsson, K.; Ingthorsson, O.; Gudmundsson, V.; Jonsdottir, I.; Halldorsson, G.H. Genome-wide association identifies seven loci for pelvic organ prolapse in Iceland and the UK Biobank. Commun. Biol. 2020, 3, 129. [Google Scholar] [CrossRef]
  42. Pujol-Gualdo, N.; Läll, K.; Lepamets, M.; Estonian Biobank Research Team; Rossi, H.-R.; Arffman, R.K.; Piltonen, T.T.; Mägi, R. Advancing our understanding of genetic risk factors and potential personalized strategies for pelvic organ prolapse. Nat. Commun. 2022, 13, 3584. [Google Scholar] [CrossRef] [PubMed]
  43. Matsunami, M.; Imamura, M.; Ashikari, A.; Liu, X.; Tomizuka, K.; Hikino, K.; Miwa, K.; Kadekawa, K.; Suda, T. Genome-wide association studies for pelvic organ prolapse in the Japanese population. Commun. Biol. 2024, 7, 1188. [Google Scholar] [CrossRef] [PubMed]
  44. Allen-Brady, K.; Cannon-Albright, L.; Farnham, J.M.; Teerlink, C.; Vierhout, M.E.; van Kempen, L.C.; Kluivers, K.B.; Norton, P.A. Identification of six loci associated with pelvic organ prolapse using genome-wide association analysis. Obstet. Gynecol. 2011, 118, 1345–1353. [Google Scholar] [CrossRef] [PubMed]
  45. Gabriel, B.; Denschlag, D.; Göbel, H.; Fittkow, C.; Werner, M.; Gitsch, G.; Watermann, D. Uterosacral ligament in postmenopausal women with or without pelvic organ prolapse. Int. Urogynecol. J. 2005, 16, 475–479. [Google Scholar] [CrossRef] [PubMed]
  46. Skorupski, P.; Król, J.; Staręga, J.; Adamiak, A.; Jankiewicz, K.; Rechberger, T. An α-1 chain of type I collagen Sp1-binding site polymorphism in women suffering from stress urinary incontinence. Am. J. Obstet. Gynecol. 2006, 194, 346–350. [Google Scholar] [CrossRef] [PubMed]
  47. Steffensen, L.B.; Rasmussen, L.M. A role for collagen type IV in cardiovascular disease? Am. J. Physiol.-Heart Circ. Physiol. 2018, 315, H610–H625. [Google Scholar] [CrossRef]
  48. Seppinen, L.; Sormunen, R.; Soini, Y.; Elamaa, H.; Heljasvaara, R.; Pihlajaniemi, T. Lack of collagen XVIII accelerates cutaneous wound healing, while overexpression of its endostatin domain leads to delayed healing. Matrix Biol. 2008, 27, 535–546. [Google Scholar] [CrossRef]
  49. Ward, R.M.; Edwards, D.R.V.; Edwards, T.; Giri, A.; Jerome, R.N.; Wu, J.M. Genetic epidemiology of pelvic organ prolapse: A systematic review. Am. J. Obstet. Gynecol. 2014, 211, 326–335. [Google Scholar] [CrossRef] [PubMed]
  50. Niu, K.; Chen, X.; Lu, Y. COL3A1 rs1800255 polymorphism is associated with pelvic organ prolapse susceptibility in Caucasian individuals: Evidence from a meta-analysis. PLoS ONE 2021, 16, e0250943. [Google Scholar] [CrossRef] [PubMed]
  51. Dos Santos, R.G.; Pepicelli, F.C.; Batista, N.C.; de Carvalho, C.V.; Bortolini, M.A.; Castro, R.A. Collagen XVIII and LOXL-4 polymorphisms in women with and without advanced pelvic organ prolapse. Int. Urogynecol. J. 2018, 29, 893–898. [Google Scholar] [CrossRef] [PubMed]
  52. Giri, A.; Wu, J.M.; Ward, R.M.; Hartmann, K.E.; Park, A.J.; North, K.E.; Graff, M.; Wallace, R.B.; Bareh, G.; Qi, L. Genetic determinants of pelvic organ prolapse among African American and Hispanic women in the Women’s Health Initiative. PLoS ONE 2015, 10, e0141647. [Google Scholar] [CrossRef] [PubMed]
  53. Mecham, R.P. Elastin synthesis and fiber assembly. Ann. N. Y. Acad. Sci. 1991, 624, 137–146. [Google Scholar] [CrossRef]
  54. Ozsvar, J.; Yang, C.; Cain, S.A.; Baldock, C.; Tarakanova, A.; Weiss, A.S. Tropoelastin and elastin assembly. Front. Bioeng. Biotechnol. 2021, 9, 643110. [Google Scholar] [CrossRef]
  55. Wise, S.G.; Weiss, A.S. Tropoelastin. Int. J. Biochem. Cell Biol. 2009, 41, 494–497. [Google Scholar] [CrossRef] [PubMed]
  56. Drewes, P.G.; Yanagisawa, H.; Starcher, B.; Hornstra, I.; Csiszar, K.; Marinis, S.I.; Keller, P.; Word, R.A. Pelvic organ prolapse in fibulin-5 knockout mice: Pregnancy-induced changes in elastic fiber homeostasis in mouse vagina. Am. J. Pathol. 2007, 170, 578–589. [Google Scholar] [CrossRef] [PubMed]
  57. Cox, C.K.; Pandit, A.; Zawistowski, M.; Dutta, D.; Narla, G.; Swenson, C.W. Genome-Wide Association Study of Pelvic Organ Prolapse Using the Michigan Genomics Initiative. Urogynecology 2021, 27, 502–506. [Google Scholar] [CrossRef]
  58. Zhao, B.h.; Zhou, J.h. Decreased expression of elastin, fibulin-5 and lysyl oxidase-like 1 in the uterosacral ligaments of postmenopausal women with pelvic organ prolapse. J. Obstet. Gynaecol. Res. 2012, 38, 925–931. [Google Scholar] [CrossRef] [PubMed]
  59. Allen-Brady, K.; Cannon-Albright, L.A.; Farnham, J.M.; Norton, P.A. Evidence for pelvic organ prolapse predisposition genes on chromosomes 10 and 17. Am. J. Obs. Gynecol. 2015, 212, e771–e777. [Google Scholar] [CrossRef]
  60. Pulst, S.M. Genetic linkage analysis. Arch. Neurol. 1999, 56, 667–672. [Google Scholar] [CrossRef] [PubMed]
  61. Liu, X.; Zhao, Y.; Gao, J.; Pawlyk, B.; Starcher, B.; Spencer, J.A.; Yanagisawa, H.; Zuo, J.; Li, T. Elastic fiber homeostasis requires lysyl oxidase–like 1 protein. Nat. Genet. 2004, 36, 178–182. [Google Scholar] [CrossRef] [PubMed]
  62. Nikolova, G.; Lee, H.; Berkovitz, S.; Nelson, S.; Sinsheimer, J.; Vilain, E.; Rodríguez, L.V. Sequence variant in the laminin γ1 (LAMC1) gene associated with familial pelvic organ prolapse. Hum. Genet. 2007, 120, 847–856. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, C.; Hill, L.D.; Schubert, C.M.; Strauss III, J.F.; Matthews, C.A. Is laminin γ-1 a candidate gene for advanced pelvic organ prolapse? Am. J. Obstet. Gynecol. 2010, 202, 505.e501–505.e505. [Google Scholar] [CrossRef]
  64. Wu, J.M.; Visco, A.G.; Grass, E.A.; Craig, D.M.; Fulton, R.G.; Haynes, C.; Amundsen, C.L.; Shah, S.H. Comprehensive analysis of LAMC1 genetic variants in advanced pelvic organ prolapse. Am. J. Obstet. Gynecol. 2012, 206, 447.e441–447.e446. [Google Scholar] [CrossRef] [PubMed]
  65. Nakad, B.; Fares, F.; Azzam, N.; Feiner, B.; Zilberlicht, A.; Abramov, Y. Estrogen receptor and laminin genetic polymorphism among women with pelvic organ prolapse. Taiwan. J. Obstet. Gynecol. 2017, 56, 750–754. [Google Scholar] [CrossRef] [PubMed]
  66. Murphy, G. Tissue inhibitors of metalloproteinases. Genome Biol. 2011, 12, 233. [Google Scholar] [CrossRef] [PubMed]
  67. Fadista, J.; Skotte, L.; Karjalainen, J.; Abner, E.; Sørensen, E.; Ullum, H.; Werge, T.; iPSYCH Group; Esko, T.; Milani, L.; et al. Comprehensive genome-wide association study of different forms of hernia identifies more than 80 associated loci. Nat. Commun. 2022, 13, 3200. [Google Scholar] [CrossRef]
  68. Xu, K.; Sun, Y.; Al-Ani, M.K.; Wang, C.; Sha, Y.; Sung, K.P.; Dong, N.; Qiu, X.; Yang, L. Synergistic promoting effects of bone morphogenetic protein 12/connective tissue growth factor on functional differentiation of tendon derived stem cells and patellar tendon window defect regeneration. J. Biomech. 2018, 66, 95–102. [Google Scholar] [CrossRef]
  69. Haddad-Weber, M.; Prager, P.; Kunz, M.; Seefried, L.; Jakob, F.; Murray, M.M.; Evans, C.H.; Nöth, U.; Steinert, A.F. BMP12 and BMP13 gene transfer induce ligamentogenic differentiation in mesenchymal progenitor and anterior cruciate ligament cells. Cytotherapy 2010, 12, 505–513. [Google Scholar] [CrossRef] [PubMed]
  70. Wei, J.; Attaar, M.; Shi, Z.; Na, R.; Resurreccion, W.; Haggerty, S.; Zheng, S.; Helfand, B.; Ujiki, M.; Xu, J. Identification of fifty-seven novel loci for abdominal wall hernia development and their biological and clinical implications: Results from the UK Biobank. Hernia 2022, 26, 335–348. [Google Scholar] [CrossRef] [PubMed]
  71. Connell, K.A.; Guess, M.K.; Chen, H.; Andikyan, V.; Bercik, R.; Taylor, H.S. HOXA11 is critical for development and maintenance of uterosacral ligaments and deficient in pelvic prolapse. J. Clin. Investig. 2008, 118, 1050–1055. [Google Scholar] [CrossRef] [PubMed]
  72. Ma, Y.; Guess, M.; Datar, A.; Hennessey, A.; Cardenas, I.; Johnson, J.; Connell, K.A. Knockdown of Hoxa11 in vivo in the uterosacral ligament and uterus of mice results in altered collagen and matrix metalloproteinase activity. Biol. Reprod. 2012, 86, 100. [Google Scholar] [CrossRef] [PubMed]
  73. Castelán, F.; Cuevas-Romero, E.; Martínez-Gómez, M. The expression of hormone receptors as a gateway toward understanding endocrine actions in female pelvic floor muscles. Endocr. Metab. Immune Disord.-Drug Targets (Former. Curr. Drug Targets-Immune Endocr. Metab. Disord.) 2020, 20, 305–320. [Google Scholar] [CrossRef] [PubMed]
  74. Koehler, K.F.; Helguero, L.A.; Haldosén, L.-A.; Warner, M.; Gustafsson, J.-A.k. Reflections on the discovery and significance of estrogen receptor β. Endocr. Rev. 2005, 26, 465–478. [Google Scholar] [CrossRef]
  75. Chen, H.-Y.; Chung, Y.-W.; Lin, W.-Y.; Chen, W.-C.; Tsai, F.-J.; Tsai, C.-H. Estrogen receptor α polymorphism is associated with pelvic organ prolapse risk. Int. Urogynecol. J. 2008, 19, 1159–1163. [Google Scholar] [CrossRef]
  76. Chen, H.-Y.; Chung, Y.-W.; Lin, W.-Y.; Chen, W.-C.; Tsai, F.-J.; Tsai, C.-H. Progesterone receptor polymorphism is associated with pelvic organ prolapse risk. Acta Obstet. Gynecol. Scand. 2009, 88, 835–838. [Google Scholar] [CrossRef]
  77. Hall, J.M.; Couse, J.F.; Korach, K.S. The multifaceted mechanisms of estradiol and estrogen receptor signaling. J. Biol. Chem. 2001, 276, 36869–36872. [Google Scholar] [CrossRef]
  78. Alperin, M.; Burnett, L.; Lukacz, E.; Brubaker, L. The mysteries of menopause and urogynecologic health: Clinical and scientific gaps. Menopause 2019, 26, 103–111. [Google Scholar] [CrossRef] [PubMed]
  79. Chen, H.-Y.; Wan, L.; Chung, Y.-W.; Chen, W.-C.; Tsai, F.-J.; Tsai, C.-H. Estrogen receptor β gene haplotype is associated with pelvic organ prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 138, 105–109. [Google Scholar] [CrossRef]
  80. Sollis, E.; Mosaku, A.; Abid, A.; Buniello, A.; Cerezo, M.; Gil, L.; Groza, T.; Güneş, O.; Hall, P.; Hayhurst, J. The NHGRI-EBI GWAS Catalog: Knowledgebase and deposition resource. Nucleic Acids Res. 2023, 51, D977–D985. [Google Scholar] [CrossRef]
  81. Sapkota, Y.; Steinthorsdottir, V.; Morris, A.P.; Fassbender, A.; Rahmioglu, N.; De Vivo, I.; Buring, J.E.; Zhang, F.; Edwards, T.L.; Jones, S. Meta-analysis identifies five novel loci associated with endometriosis highlighting key genes involved in hormone metabolism. Nat. Commun. 2017, 8, 15539. [Google Scholar] [CrossRef] [PubMed]
  82. Biason-Lauber, A.; Konrad, D. WNT4 and sex development. Sex. Dev. 2008, 2, 210–218. [Google Scholar] [CrossRef] [PubMed]
  83. Mandel, H.; Shemer, R.; Borochowitz, Z.U.; Okopnik, M.; Knopf, C.; Indelman, M.; Drugan, A.; Tiosano, D.; Gershoni-Baruch, R.; Choder, M. SERKAL syndrome: An autosomal-recessive disorder caused by a loss-of-function mutation in WNT4. Am. J. Hum. Genet. 2008, 82, 39–47. [Google Scholar] [CrossRef]
  84. Sherry, S.T.; Ward, M.-H.; Kholodov, M.; Baker, J.; Phan, L.; Smigielski, E.M.; Sirotkin, K. dbSNP: The NCBI database of genetic variation. Nucleic Acids Res. 2001, 29, 308–311. [Google Scholar] [CrossRef] [PubMed]
  85. O’Leary, N.A.; Wright, M.W.; Brister, J.R.; Ciufo, S.; Haddad, D.; McVeigh, R.; Rajput, B.; Robbertse, B.; Smith-White, B.; Ako-Adjei, D. Reference sequence (RefSeq) database at NCBI: Current status, taxonomic expansion, and functional annotation. Nucleic Acids Res. 2016, 44, D733–D745. [Google Scholar] [CrossRef] [PubMed]
  86. Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [PubMed]
  87. Filomeni, G.; De Zio, D.; Cecconi, F. Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 2015, 22, 377–388. [Google Scholar] [CrossRef]
  88. Hong, S.; Hong, L.; Wu, D.; Li, B.; Liu, C.; Guo, W.; Min, J.; Hu, M.; Zhao, Y.; Yang, Q. Oxidative damage to human parametrial ligament fibroblasts induced by mechanical stress. Mol. Med. Rep. 2015, 12, 5342–5348. [Google Scholar] [CrossRef] [PubMed]
  89. Kim JiYoung, K.J.; Kim EunJae, K.E.; Jeon MyungJae, J.M.; Kim Ran, K.R.; Lee MinWoo, L.M.; Kim SuhngWook, K.S. Association between susceptibility to advanced pelvic organ prolapse and glutathione S-transferase P1 Ile105Val polymorphism. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014, 175, 205–208. [Google Scholar] [CrossRef]
  90. Kim, J.Y.; Kim, E.J.; Jeon, M.J.; Kim, H.; Moon, Y.J.; Bai, S.W. Association between the poly (ADP-ribose) polymerase-1 gene polymorphism and advanced pelvic organ prolapse. Menopause 2014, 21, 177–181. [Google Scholar] [CrossRef] [PubMed]
  91. Luo, X.; Kraus, W.L. On PAR with PARP: Cellular stress signaling through poly (ADP-ribose) and PARP-1. Genes Dev. 2012, 26, 417–432. [Google Scholar] [CrossRef] [PubMed]
  92. Von Scheidt, M.; Zhao, Y.; de Aguiar Vallim, T.Q.; Che, N.; Wierer, M.; Seldin, M.M.; Franzén, O.; Kurt, Z.; Pang, S.; Bongiovanni, D. Transcription factor MAFF (MAF basic leucine zipper transcription factor F) regulates an atherosclerosis relevant network connecting inflammation and cholesterol metabolism. Circulation 2021, 143, 1809–1823. [Google Scholar] [CrossRef]
  93. Darwich, R.; Li, W.; Yamak, A.; Komati, H.; Andelfinger, G.; Sun, K.; Nemer, M. KLF13 is a genetic modifier of the Holt-Oram syndrome gene TBX5. Hum. Mol. Genet. 2017, 26, 942–954. [Google Scholar] [CrossRef] [PubMed]
  94. Fukuda, R.; Gunawan, F.; Ramadass, R.; Beisaw, A.; Konzer, A.; Mullapudi, S.T.; Gentile, A.; Maischein, H.-M.; Graumann, J.; Stainier, D.Y. Mechanical forces regulate cardiomyocyte myofilament maturation via the VCL-SSH1-CFL axis. Dev. Cell 2019, 51, 62–77.e5. [Google Scholar] [CrossRef] [PubMed]
  95. Fitz, F.F.; Bortolini, M.A.T.; Pereira, G.M.V.; Salerno, G.R.F.; Castro, R.A. PEOPLE: Lifestyle and comorbidities as risk factors for pelvic organ prolapse—A systematic review and meta-analysis PEOPLE: PElvic Organ Prolapse Lifestyle comorbiditiEs. Int. Urogynecol. J. 2023, 34, 2007–2032. [Google Scholar] [CrossRef] [PubMed]
  96. Isık, H.; Aynıoglu, O.; Sahbaz, A.; Selimoglu, R.; Timur, H.; Harma, M. Are hypertension and diabetes mellitus risk factors for pelvic organ prolapse? Eur. J. Obstet. Gynecol. Reprod. Biol. 2016, 197, 59–62. [Google Scholar] [CrossRef]
  97. Rodrigues, A.M.; Girão, M.J.B.C.; da Silva, I.D.C.G.; Sartori, M.G.F.; Martins, K.d.F.; Castro, R.d.A. COL1A1 Sp1-binding site polymorphism as a risk factor for genital prolapse. Int. Urogynecol. J. 2008, 19, 1471–1475. [Google Scholar] [CrossRef] [PubMed]
  98. Cho, H.J.; Jung, H.J.; Kim, S.K.; Choi, J.R.; Cho, N.H.; Bai, S.W. Polymorphism of a COLIA1 gene Sp1 binding site in Korean women with pelvic organ prolapse. Yonsei Med. J. 2009, 50, 564–568. [Google Scholar] [CrossRef] [PubMed]
  99. Feiner, B.; Fares, F.; Azam, N.; Auslender, R.; David, M.; Abramov, Y. Does COLIA1 SP1-binding site polymorphism predispose women to pelvic organ prolapse? Int. Urogynecol. J. 2009, 20, 1061–1065. [Google Scholar] [CrossRef]
  100. Batista, N.C.; Bortolini, M.A.; Silva, R.S.; Teixeira, J.B.; Melo, N.C.; Santos, R.G.; Pepicelli, F.A.; Castro, R.A. Collagen I and collagen III polymorphisms in women with pelvic organ prolapse. Neurourol. Urodyn. 2020, 39, 1977–1984. [Google Scholar] [CrossRef]
  101. Palos, C.C.; Timm, B.F.; de Souza Paulo, D.; Fernandes, C.E.; de Souto, R.P.; Oliveira, E. Evaluation of COLIA1-1997 G/T polymorphism as a related factor to genital prolapse. Int. Urogynecol. J. 2020, 31, 133–137. [Google Scholar] [CrossRef] [PubMed]
  102. de Freitas Rosa, J.P.; Haddad, R.F.; Maeda, F.G.R.; Souto, R.P.; Fernandes, C.E.; de Oliveira, E. Association between col1a2 polymorphism and the occurrence of pelvic organ prolapse in Brazilian women. Rev. Bras. De Ginecol. E Obs./RBGO Gynecol. Obstet. 2019, 41, 031–036. [Google Scholar] [CrossRef]
  103. Teixeira, F.H.; Fernandes, C.E.; do Souto, R.P.; de Oliveira, E. Polymorphism rs1800255 from COL3A1 gene and the risk for pelvic organ prolapse. Int. Urogynecol. J. 2020, 31, 73–78. [Google Scholar] [CrossRef]
  104. Paula, M.V.B.d.; Lira Júnior, M.A.d.F.; Souto, R.P.; Fernandes, C.E.; Oliveira, E.d. Evaluation of the fibulin 5 gene polymorphism as a factor related to the occurrence of pelvic organ prolapse. Rev. Da Assoc. Médica Bras. 2020, 66, 680–686. [Google Scholar] [CrossRef]
  105. Neupane, R.; Sadeghi, Z.; Fu, R.; Hagstrom, S.A.; Moore, C.K.; Daneshgari, F. Mutation screen of LOXL1 in patients with female pelvic organ prolapse. Urogynecology 2014, 20, 316–321. [Google Scholar] [CrossRef]
  106. Skorupski, P.; Jankiewicz, K.; Miotła, P.; Marczak, M.; Kulik-Rechberger, B.; Rechberger, T. The polymorphisms of the MMP-1 and the MMP-3 genes and the risk of pelvic organ prolapse. Int. Urogynecol. J. 2013, 24, 1033–1038. [Google Scholar] [CrossRef] [PubMed]
  107. Karachalios, C.; Bakas, P.; Kaparos, G.; Demeridou, S.; Liapis, I.; Grigoriadis, C.; Liapis, A. Matrix metalloproteinase-3 gene promoter polymorphisms: A potential risk factor for pelvic organ prolapse. Biomed. Rep. 2016, 5, 337–343. [Google Scholar] [CrossRef] [PubMed]
  108. Maeda, P.M.; Bicudo, A.P.S.; Watanabe, R.T.; Fonseca, T.S.; do Souto, R.P.; Fernandes, C.E.; de Oliveira, E. Study of the polymorphism rs3025058 of the MMP-3 gene and risk of pelvic organ prolapse in Brazilian women. Eur. J. Obstet. Gynecol. Reprod. Biol. X 2019, 3, 100031. [Google Scholar] [CrossRef]
  109. Ghersel, F.R.; Souto, R.P.; Gonzales, E.W.P.; Paulo, D.S.; Fernandes, C.E.; Oliveira, E. Assessment of metalloproteinase matrix 9 (MMP9) gene polymorphisms risk factors for pelvic organ prolapse in the Brazilian population. Rev. Bras. De Ginecol. E Obs. 2019, 41, 164–169. [Google Scholar] [CrossRef]
  110. Singh, K.P.; Miaskowski, C.; Dhruva, A.A.; Flowers, E.; Kober, K.M. Mechanisms and measurement of changes in gene expression. Biol. Res. Nurs. 2018, 20, 369–382. [Google Scholar] [CrossRef] [PubMed]
  111. Battle, A.; Brown, C.D.; Engelhardt, B.E.; Montgomery, S.B. Genetic effects on gene expression across human tissues. Nature 2017, 550, 204–213. [Google Scholar] [CrossRef] [PubMed]
  112. DeLancey, J.O. Anatomie aspects of vaginal eversion after hysterectomy. Am. J. Obstet. Gynecol. 1992, 166, 1717–1728. [Google Scholar] [CrossRef]
  113. Zhao, Y.; Xia, Z.; Lin, T.; Qin, M. Transforming growth factor β 1 and p44/42 expression in cardinal ligament tissues of patients with pelvic organ prolapse. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2021, 27, e930433-1. [Google Scholar] [CrossRef]
  114. Yucel, N.; Usta, A.; Guzin, K.; Kanter, M.; Bilgic, E.; Ozel, N.O.; Ozgul, M. Immunohistochemical analysis of connective tissue in patients with pelvic organ prolapse. J. Mol. Histol. 2013, 44, 97–102. [Google Scholar] [CrossRef]
  115. Dökmeci, F.; Tekşen, F.; Çetinkaya, Ş.E.; Özkan, T.; Kaplan, F.; Köse, K. Expressions of homeobox, collagen and estrogen genes in women with uterine prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2019, 233, 26–29. [Google Scholar] [CrossRef] [PubMed]
  116. Zhu, Y.-p.; Xie, T.; Guo, T.; Sun, Z.-j.; Zhu, L.; Lang, J.-h. Evaluation of extracellular matrix protein expression and apoptosis in the uterosacral ligaments of patients with or without pelvic organ prolapse. Int. Urogynecol. J. 2021, 32, 2273–2281. [Google Scholar] [CrossRef] [PubMed]
  117. Chen, S.; Zheng, Q.; Zhang, L.; Chen, L.; Wang, J. Effect of Vaginal Microecological Alterations on Female Pelvic Organ Prolapse. Int. Urogynecol. J. 2024, 35, 881–891. [Google Scholar] [CrossRef] [PubMed]
  118. Söderberg, M.W.; Byström, B.; Kalamajski, S.; Malmström, A.; Ekman-Ordeberg, G. Gene expressions of small leucine-rich repeat proteoglycans and fibulin-5 are decreased in pelvic organ prolapse. MHR Basic Sci. Reprod. Med. 2009, 15, 251–257. [Google Scholar] [CrossRef]
  119. Moalli, P.A.; Shand, S.H.; Zyczynski, H.M.; Gordy, S.C.; Meyn, L.A. Remodeling of vaginal connective tissue in patients with prolapse. Obstet. Gynecol. 2005, 106, 953–963. [Google Scholar] [CrossRef]
  120. Lin, S.-Y.; Tee, Y.-T.; Ng, S.-C.; Chang, H.; Lin, P.; Chen, G.-D. Changes in the extracellular matrix in the anterior vagina of women with or without prolapse. Int. Urogynecol. J. 2007, 18, 43–48. [Google Scholar] [CrossRef]
  121. Mosier, E.; Lin, V.K.; Zimmern, P. Extracellular matrix expression of human prolapsed vaginal wall. Neurourol. Urodyn. Off. J. Int. Cont. Soc. 2010, 29, 582–586. [Google Scholar] [CrossRef] [PubMed]
  122. Zhou, L.; Lee, J.H.; Wen, Y.; Constantinou, C.; Yoshinobu, M.; Omata, S.; Chen, B. Biomechanical properties and associated collagen composition in vaginal tissue of women with pelvic organ prolapse. J. Urol. 2012, 188, 875–880. [Google Scholar] [CrossRef]
  123. Kerkhof, M.H.; Ruiz-Zapata, A.M.; Bril, H.; Bleeker, M.C.; Belien, J.A.; Stoop, R.; Helder, M.N. Changes in tissue composition of the vaginal wall of premenopausal women with prolapse. Am. J. Obstet. Gynecol. 2014, 210, 168.e161–168.e169. [Google Scholar] [CrossRef] [PubMed]
  124. Vetuschi, A.; D’Alfonso, A.; Sferra, R.; Zanelli, D.; Pompili, S.; Patacchiola, F.; Gaudio, E.; Carta, G. Changes in muscularis propria of anterior vaginal wall in women with pelvic organ prolapse. Eur. J. Histochem. EJH 2016, 60, 2604. [Google Scholar] [CrossRef] [PubMed]
  125. Hu, Y.; Wu, R.; Li, H.; Gu, Y.; Wei, W. Expression and significance of metalloproteinase and collagen in vaginal wall tissues of patients with pelvic organ prolapse. Ann. Clin. Lab. Sci. 2017, 47, 698–705. [Google Scholar]
  126. Vetuschi, A.; Pompili, S.; Gallone, A.; D’Alfonso, A.; Carbone, M.G.; Carta, G.; Festuccia, C.; Gaudio, E.; Colapietro, A.; Sferra, R. Immunolocalization of advanced glycation end products, mitogen activated protein kinases, and transforming growth factor-β/Smads in pelvic organ prolapse. J. Histochem. Cytochem. 2018, 66, 673–686. [Google Scholar] [CrossRef]
  127. Lin, T.; Ji, Y.; Zhao, Y.; Xia, Z. Expression of COX-2 and Nrf2/GPx3 in the anterior vaginal wall tissues of women with pelvic organ prolapse. Arch. Gynecol. Obstet. 2021, 303, 1245–1253. [Google Scholar] [CrossRef] [PubMed]
  128. Goepel, C. Differential elastin and tenascin immunolabeling in the uterosacral ligaments in postmenopausal women with and without pelvic organ prolapse. Acta Histochem. 2008, 110, 204–209. [Google Scholar] [CrossRef]
  129. de Landsheere, L.; Blacher, S.; Munaut, C.; Nusgens, B.; Rubod, C.; Noel, A.; Foidart, J.-M.; Cosson, M.; Nisolle, M. Changes in elastin density in different locations of the vaginal wall in women with pelvic organ prolapse. Int. Urogynecol. J. 2014, 25, 1673–1681. [Google Scholar] [CrossRef]
  130. Takacs, P.; Nassiri, M.; Candiotti, K.; Yang, J.; Yavagal, S.; Medina, C.A. Differential expression of fibulins in the uterosacral ligaments of women with uterine prolapse. Arch. Gynecol. Obstet. 2010, 282, 389–394. [Google Scholar] [CrossRef]
  131. Ahn, K.; Kim, T.; Hur, J.; Kim, S.; Lee, K.; Kim, Y. Relationship between the expression of fibulin-3 and anterior vaginal wall prolapse. J. Obstet. Gynaecol. 2012, 32, 362–366. [Google Scholar] [CrossRef] [PubMed]
  132. Zhou, Y.; Ling, O.; Bo, L. Expression and significance of lysyl oxidase-like 1 and fibulin-5 in the cardinal ligament tissue of patients with pelvic floor dysfunction. J. Biomed. Res. 2013, 27, 23. [Google Scholar] [CrossRef]
  133. Klutke, J.; Ji, Q.; Campeau, J.; Starcher, B.; Carlos Felix, J.; Stanczyk, F.Z.; Klutke, C. Decreased endopelvic fascia elastin content in uterine prolapse. Acta Obstet. Gynecol. Scand. 2008, 87, 111–115. [Google Scholar] [CrossRef] [PubMed]
  134. Jung, H.J.; Jeon, M.J.; Yim, G.W.; Kim, S.K.; Choi, J.R.; Bai, S.W. Changes in expression of fibulin-5 and lysyl oxidase-like 1 associated with pelvic organ prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2009, 145, 117–122. [Google Scholar] [CrossRef]
  135. Takacs, P.; Nassiri, M.; Viciana, A.; Candiotti, K.; Fornoni, A.; Medina, C.A. Fibulin-5 expression is decreased in women with anterior vaginal wall prolapse. Int. Urogynecol. J. 2009, 20, 207–211. [Google Scholar] [CrossRef] [PubMed]
  136. Alarab, M.; Bortolini, M.A.; Drutz, H.; Lye, S.; Shynlova, O. LOX family enzymes expression in vaginal tissue of premenopausal women with severe pelvic organ prolapse. Int. Urogynecol. J. 2010, 21, 1397–1404. [Google Scholar] [CrossRef] [PubMed]
  137. Strinic, T.; Vulic, M.; Tomic, S.; Capkun, V.; Stipic, I.; Alujevic, I. Matrix metalloproteinases-1,-2 expression in uterosacral ligaments from women with pelvic organ prolapse. Maturitas 2009, 64, 132–135. [Google Scholar] [CrossRef] [PubMed]
  138. Vulic, M.; Strinic, T.; Tomic, S.; Capkun, V.; Jakus, I.A.; Ivica, S. Difference in expression of collagen type I and matrix metalloproteinase-1 in uterosacral ligaments of women with and without pelvic organ prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2011, 155, 225–228. [Google Scholar] [CrossRef]
  139. Dviri, M.; Leron, E.; Dreiher, J.; Mazor, M.; Shaco-Levy, R. Increased matrix metalloproteinases-1,-9 in the uterosacral ligaments and vaginal tissue from women with pelvic organ prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2011, 156, 113–117. [Google Scholar] [CrossRef]
  140. Usta, A.; Guzin, K.; Kanter, M.; Ozgül, M.; Usta, C.S. Expression of matrix metalloproteinase-1 in round ligament and uterosacral ligament tissue from women with pelvic organ prolapse. J. Mol. Histol. 2014, 45, 275–281. [Google Scholar] [CrossRef] [PubMed]
  141. Alarab, M.; Kufaishi, H.; Lye, S.; Drutz, H.; Shynlova, O. Expression of extracellular matrix-remodeling proteins is altered in vaginal tissue of premenopausal women with severe pelvic organ prolapse. Reprod. Sci. 2014, 21, 704–715. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, X.; Li, Y.; Chen, J.; Guo, X.; Guan, H.; Li, C. Differential expression profiling of matrix metalloproteinases and tissue inhibitors of metalloproteinases in females with or without pelvic organ prolapse. Mol. Med. Rep. 2014, 10, 2004–2008. [Google Scholar] [CrossRef]
  143. Moon, Y.J.; Choi, J.R.; Jeon, M.J.; Kim, S.K.; Bai, S.W. Alteration of elastin metabolism in women with pelvic organ prolapse. J. Urol. 2011, 185, 1786–1792. [Google Scholar] [CrossRef]
  144. Leegant, A.; Zuckerwise, L.C.; Downing, K.; Brouwer-Visser, J.; Zhu, C.; Cossio, M.J.; Strube, F.; Xie, X.; Banks, E.; Huang, G.S. Transforming growth factor β1 and extracellular matrix protease expression in the uterosacral ligaments of patients with and without pelvic organ prolapse. Urogynecology 2015, 21, 53–58. [Google Scholar] [CrossRef] [PubMed]
  145. Liang, C.-C.; Huang, H.-Y.; Tseng, L.-H.; Chang, S.-D.; Lo, T.-S.; Lee, C.-L. Expression of matrix metalloproteinase-2 and tissue inhibitors of metalloproteinase-1 (TIMP-1, TIMP-2 and TIMP-3) in women with uterine prolapse but without urinary incontinence. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010, 153, 94–98. [Google Scholar] [CrossRef] [PubMed]
  146. Bai, S.W.; Chung, D.J.; Yoon, J.M.; Shin, J.S.; Kim, S.K.; Park, K.H. Roles of estrogen receptor, progesterone receptor, p53 and p21 in pathogenesis of pelvic organ prolapse. Int. Urogynecol. J. 2005, 16, 492–496. [Google Scholar] [CrossRef] [PubMed]
  147. Dietrich, W.; Elenskaia, K.; Obermayr, E.; Horvat, R.; Mayerhofer, K.; Umek, W.; Zeillinger, R.; Hanzal, E. Relaxin and gonadal steroid receptors in uterosacral ligaments of women with and without pelvic organ prolapse. Int. Urogynecol. J. 2012, 23, 495–500. [Google Scholar] [CrossRef]
  148. Sun, M.-J.; Cheng, Y.-S.; Sun, R.; Cheng, W.-L. Changes in mitochondrial DNA copy number and extracellular matrix (ECM) proteins in the uterosacral ligaments of premenopausal women with pelvic organ prolapse. Taiwan. J. Obstet. Gynecol. 2016, 55, 9–15. [Google Scholar] [CrossRef]
  149. Tola, E.N.; Koroglu, N.; Yıldırım, G.Y.; Koca, H.B. The role of ADAMTS-2, collagen type-1, TIMP-3 and papilin levels of uterosacral and cardinal ligaments in the etiopathogenesis of pelvic organ prolapse among women without stress urinary incontinence. Eur. J. Obstet. Gynecol. Reprod. Biol. 2018, 231, 158–163. [Google Scholar] [CrossRef]
  150. Putra, I.G.M.; Warsita, I.G.N.; Suwiyoga, K.; Manuaba, I.G.F.; Budiana, I.N.G.; Wiradnyana, A.G.P. Low expression of collagen type-1 in sacrouterine ligament as risk factor of stage III–IV uterine prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2020, 249, 32–36. [Google Scholar] [CrossRef] [PubMed]
  151. Chen, Y.; Huang, J.; Hu, C.; Hua, K. Relationship of advanced glycation end products and their receptor to pelvic organ prolapse. Int. J. Clin. Exp. Pathol. 2015, 8, 2288. [Google Scholar]
  152. Eser, A.; Unlubilgin, E.; Hizli, F.; Acar, M.; Kamalak, Z.; Kosus, A.; Kosus, N.; Hizli, D.; Gunduz, E. Is there a relationship between pelvic organ prolapse and tissue Fibrillin-1 levels? Int. Neurourol. J. 2015, 19, 164. [Google Scholar] [CrossRef] [PubMed]
  153. Bodner-Adler, B.; Bodner, K.; Kimberger, O.; Halpern, K.; Schneidinger, C.; Haslinger, P.; Schneeberger, C.; Horvat, R.; Umek, W. The role of tenascin-X in the uterosacral ligaments of postmenopausal women with pelvic organ prolapse: An immunohistochemical study. Int. Urogynecol. J. 2020, 31, 101–106. [Google Scholar] [CrossRef]
  154. Choy, K.; Liu, Y.; Chu, C.; Wang, C.; Lui, W.; Lee, L.; Pang, M.; Rogers, M.; Yip, S. High isoprostane level in cardinal ligament-derived fibroblasts and urine sample of women with uterine prolapse. BJOG Int. J. Obstet. Gynaecol. 2008, 115, 1179–1183. [Google Scholar] [CrossRef]
  155. Yılmaz, N.; Ozaksit, G.; Terzi, Y.K.; Yılmaz, S.; Budak, B.; Aksakal, O.; Şahin, F.İ. HOXA11 and MMP2 gene expression in uterosacral ligaments of women with pelvic organ prolapse. J. Turk. Ger. Gynecol. Assoc. 2014, 15, 104. [Google Scholar] [CrossRef]
  156. Connell, K.A.; Guess, M.K.; Tate, A.; Andikyan, V.; Bercik, R.; Taylor, H.S. Diminished vaginal HOXA13 expression in women with pelvic organ prolapse. Menopause 2009, 16, 529–533. [Google Scholar] [CrossRef]
  157. Fang, G.; Hong, L.; Liu, C.; Yang, Q.; Zhang, Q.; Li, Y.; Li, B.; Wu, D.; Wu, W.; Shi, H. Oxidative status of cardinal ligament in pelvic organ prolapse. Exp. Ther. Med. 2018, 16, 3293–3302. [Google Scholar] [CrossRef] [PubMed]
  158. Kim, E.J.; Chung, N.; Park, S.H.; Lee, K.-H.; Kim, S.W.; Kim, J.Y.; Bai, S.W.; Jeon, M.J. Involvement of oxidative stress and mitochondrial apoptosis in the pathogenesis of pelvic organ prolapse. J. Urol. 2013, 189, 588–594. [Google Scholar] [CrossRef] [PubMed]
  159. Jakus, I.A.; Jakus, D.; Aračić, N.; Stipić, I.; Vilović, K. Immunohistochemical expression of hypoxia-inducible factor-1α in stromal cells of vaginal tissue in post-menopausal women with pelvic organ prolapse. Indian J. Med. Res. 2017, 146, S63–S67. [Google Scholar] [CrossRef]
  160. Sun, M.-J.; Cheng, Y.-S.; Liu, C.-S.; Sun, R. Changes in the PGC-1α and mtDNA copy number may play a role in the development of pelvic organ prolapse in pre-menopausal patients. Taiwan. J. Obstet. Gynecol. 2019, 58, 526–530. [Google Scholar] [CrossRef] [PubMed]
  161. Zhu, Y.; Huang, Y.; Tan, Y.; Zhao, W.; Tian, Q. Single-cell RNA sequencing in hematological diseases. Proteomics 2020, 20, 1900228. [Google Scholar] [CrossRef] [PubMed]
  162. Li, Y.; Zhang, Q.-Y.; Sun, B.-F.; Ma, Y.; Zhang, Y.; Wang, M.; Ma, C.; Shi, H.; Sun, Z.; Chen, J. Single-cell transcriptome profiling of the vaginal wall in women with severe anterior vaginal prolapse. Nat. Commun. 2021, 12, 87. [Google Scholar] [CrossRef]
  163. Fan, W.; Wu, D.; Zhang, L.; Ye, J.; Guan, J.; Yang, Y.; Mei, X.; Chen, R. Single-cell transcriptomic data reveal the increase in extracellular matrix organization and antigen presentation abilities of fibroblasts and smooth muscle cells in patients with pelvic organ prolapse. Int. Urogynecol. J. 2023, 34, 2529–2537. [Google Scholar] [CrossRef]
  164. Miao, Y.; Wen, J.; Wang, L.; Wen, Q.; Cheng, J.; Zhao, Z.; Wu, J. scRNA-seq reveals aging-related immune cell types and regulators in vaginal wall from elderly women with pelvic organ prolapse. Front. Immunol. 2023, 14, 1084516. [Google Scholar] [CrossRef]
  165. Liu, X.; Su, M.; Wei, L.; Zhang, J.; Wang, W.; Hao, Q.; Lin, X.; Wang, L. Single-cell analysis of uterosacral ligament revealed cellular heterogeneity in women with pelvic organ prolapse. Commun. Biol. 2024, 7, 159. [Google Scholar] [CrossRef] [PubMed]
  166. Choi, S.W.; Mak, T.S.-H.; O’Reilly, P.F. Tutorial: A guide to performing polygenic risk score analyses. Nat. Protoc. 2020, 15, 2759–2772. [Google Scholar] [CrossRef] [PubMed]
  167. Eichler, E.E.; Flint, J.; Gibson, G.; Kong, A.; Leal, S.M.; Moore, J.H.; Nadeau, J.H. Missing heritability and strategies for finding the underlying causes of complex disease. Nat. Rev. Genet. 2010, 11, 446–450. [Google Scholar] [CrossRef] [PubMed]
  168. Manolio, T.A.; Collins, F.S.; Cox, N.J.; Goldstein, D.B.; Hindorff, L.A.; Hunter, D.J.; McCarthy, M.I.; Ramos, E.M.; Cardon, L.R.; Chakravarti, A. Finding the missing heritability of complex diseases. Nature 2009, 461, 747–753. [Google Scholar] [CrossRef] [PubMed]
  169. Consortium, G.P. A global reference for human genetic variation. Nature 2015, 526, 68. [Google Scholar] [CrossRef] [PubMed]
  170. Zarrei, M.; MacDonald, J.R.; Merico, D.; Scherer, S.W. A copy number variation map of the human genome. Nat. Rev. Genet. 2015, 16, 172–183. [Google Scholar] [CrossRef] [PubMed]
  171. Cavalli, G.; Heard, E. Advances in epigenetics link genetics to the environment and disease. Nature 2019, 571, 489–499. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, L.; Zheng, P.; Duan, A.; Hao, Y.; Lu, C.; Lu, D. Genome-wide DNA methylation analysis of uterosacral ligaments in women with pelvic organ prolapse. Mol. Med. Rep. 2019, 19, 391–399. [Google Scholar] [CrossRef] [PubMed]
  173. Yin, Y.; Qin, M.; Luan, M.; Xia, Z. miR-19-3p Promotes Autophagy and Apoptosis in Pelvic Organ Prolapse Through the AKT/mTOR/p70S6K Pathway: Function of miR-19-3p on Vaginal Fibroblasts by Targeting IGF-1. Urogynecology 2021, 27, e630–e638. [Google Scholar] [CrossRef] [PubMed]
  174. He, K.; Niu, G.; Gao, J.; Liu, J.X.; Qu, H. MicroRNA-92 expression may be associated with reduced estrogen receptor β1 mRNA levels in cervical portion of uterosacral ligaments in women with pelvic organ prolapse. Eur. J. Obstet. Gynecol. Reprod. Biol. 2016, 198, 94–99. [Google Scholar] [CrossRef]
Figure 1. Chromosome map of significant SNPs associated with POP. Red line: the significant SNPs identified in candidate gene association study. Blue line: the significant SNPs identified in genome-wide association study.
Figure 1. Chromosome map of significant SNPs associated with POP. Red line: the significant SNPs identified in candidate gene association study. Blue line: the significant SNPs identified in genome-wide association study.
Genes 16 00005 g001
Figure 2. Overview of POP-associated SNPs and their related gene expression in different pelvic floor tissues. Red: significant SNPs identified in candidate gene association studies. Blue: significant SNPs identified in genome-wide association studies. Abbreviations: CL: cardinal ligament; USL: uterosacral ligament; RL: round ligament; 8-OHdG: 8-hydroxy-2’ -deoxyguanosine; 4HNE: 4-Hydroxynonenal; MnSOD: manganese superoxide dismutase; GPX1: glutathione peroxidase 1; NRF-1: nuclear respiratory factor-1; NRF-2: nuclear respiratory factor-2; AGE: advanced glycation end product; COX-2: cyclooxygenase-2; PGE2: prostaglandin E2; HIF-1 α: hypoxia-inducible factors α; GPX3: glutathione peroxidase 3.
Figure 2. Overview of POP-associated SNPs and their related gene expression in different pelvic floor tissues. Red: significant SNPs identified in candidate gene association studies. Blue: significant SNPs identified in genome-wide association studies. Abbreviations: CL: cardinal ligament; USL: uterosacral ligament; RL: round ligament; 8-OHdG: 8-hydroxy-2’ -deoxyguanosine; 4HNE: 4-Hydroxynonenal; MnSOD: manganese superoxide dismutase; GPX1: glutathione peroxidase 1; NRF-1: nuclear respiratory factor-1; NRF-2: nuclear respiratory factor-2; AGE: advanced glycation end product; COX-2: cyclooxygenase-2; PGE2: prostaglandin E2; HIF-1 α: hypoxia-inducible factors α; GPX3: glutathione peroxidase 3.
Genes 16 00005 g002
Table 1. Summary of significant single nucleotide polymorphisms (SNPs) associated with the remodeling of ECM in candidate gene association studies and genome-wide association studies.
Table 1. Summary of significant single nucleotide polymorphisms (SNPs) associated with the remodeling of ECM in candidate gene association studies and genome-wide association studies.
Candidate Gene Association Studies
ComponentSNP IDAnnotationChange in NucleotideCandidate GeneEffect DirectionSignificant Association with POP *Race/EthnicitySample Size (POP/Control)Reference
Collagenrs1800255ExonicG>ACOL3A1Riskgenotype AA, OR 5.05East Asian (Chinese)84 vs. 147[29]
Riskgenotype AA, OR 5.00European (Dutch)202 vs. 102[30]
rs445348ExonicA>GCOL4A2Riskallele G, OR 2.15East Asian (Chinese)48 vs. 48[31]
rs76425569ExonicG>ACOL4A2Riskallele A, OR 2.02East Asian (Chinese)48 vs. 48[31]
rs388222IntronicC>TCOL4A2Protectiveallele T, OR 0.50East Asian (Chinese)48 vs. 48[31]
rs2281968IntronicG>ACOL4A2Riskallele A, OR 2.02East Asian (Chinese)48 vs. 48[31]
rs3827852IntronicA>GCOL5A1Protectiveallele G, OR 0.40East Asian (Chinese)48 vs. 48[31]
rs4870723ExonicA>CCOL14A1Protectiveallele C, OR 0.46East Asian (Chinese)48 vs. 48[31]
rs2305600ExonicT>CCOL14A1Protectiveallele C, OR 0.48East Asian (Chinese)48 vs. 48[31]
rs2305598ExonicT>CCOL14A1Protectiveallele C, OR 0.50East Asian (Chinese)48 vs. 48[31]
Elasticrs2018736IntronicC>AFBLN5Protectiveallele A, OR 0.73Russian210 vs. 292[32]
fibersrs12589592IntronicG>AFBLN5Protectiveallele A, OR 0.42Russian210 vs. 292[32]
Protectivegenotype AA, OR 0.11,
allele A, OR 0.48
East Asian (minority/non-minority Chinese)88 vs. 108[33]
Lysyl
oxidase
rs2862296IntergenicA>GLOXL4Riskgenotype AG, OR 3.80;
genotype GG, OR 4.50
East Asian (Japanese)52 vs. 28[34]
Lamininrs10911241IntronicA>GLAMC1Riskallele G, OR 1.71East Asian (Chinese)161 vs. 235[35]
Proteases-Upstream VariantG→GGMMP1Riskgenotype GG/GG (OR not analyzed)European (Italian)137 vs. 96[36]
rs17576ExonicA>GMMP9Riskgenotype AG, OR 5.41;
genotype GG, OR 5.77
East Asian (Chinese)92 vs. 152[37]
rs3918253IntronicC>TMMP9Riskallele T, OR 1.56Non-Hispanic White239 vs. 197[38]
rs3918256IntronicG>AMMP9Riskallele A, OR 1.56Non-Hispanic White239 vs. 197[38]
rs17435959ExonicG>CMMP10Riskgenotype GC, OR 9.59;
genotype CC, OR 4.30
East Asian (Chinese)91 vs. 172[39]
rs370850IntronicC>TADAMTS1Riskallele T, OR 3.71East Asian (Chinese)48 vs. 48[40]
rs422803IntronicC>AADAMTS1Riskallele A, OR 3.71East Asian (Chinese)48 vs. 48[40]
rs4020075’UTRC>GADAMTS1Riskallele G, OR 2.18East Asian (Chinese)48 vs. 48[40]
rs428785ExonicC>GADAMTS1Riskallele G, OR 2.18East Asian (Chinese)48 vs. 48[40]
rs434857ExonicT>GADAMTS1Riskallele G, OR 2.18East Asian (Chinese)48 vs. 48[40]
rs445784ExonicG>TADAMTS1Riskallele T, OR 2.18East Asian (Chinese)48 vs. 48[40]
rs149586801IntronicC>TADAMTS13Protectiveallele T, OR 0.18East Asian (Chinese)48 vs. 48[40]
rs2277698ExonicC>TTIMP2Protectiveallele T, OR 0.37East Asian (Chinese)48 vs. 48[40]
Genome-Wide Association Study
SNP IDBand RegionAnnotationEffect AlleleOther AlleleEAFEffect DirectionOR (95% CI)p ValueMapped Gene(s) ^Reference
rs93068942p24.13’UTRGA0.16Risk1.10 (1.08–1.12)5.61 × 10−24 GDF7[41,42]
rs14301912p16.1IntergenicTC0.48 Risk1.09 (1.06–1.12)1.00 × 10−9EFEMP1[41]
rs118998882p16.1IntronGA0.11 Risk1.11 (1.09–1.14)4.01 × 10−16EFEMP1[42]
rs37916752p16.1IntronTC0.25 Protective0.92 (0.90–0.94)1.23 × 10−13EFEMP1[41,42]
rs776481362q31.1IntergenicTG0.16 Protective0.94 (0.92–0.96)4.81 × 10−8HOXD13[42]
rs424005p15.32IntergenicGC0.36 Protective0.94 (0.92–0.96)1.65 × 10−10ADAMTS16[42]
rs108108889p22.3IntronGA0.65 Risk1.05 (1.03–1.07)4.00 × 10−8ADAMTSL1[42]
rs707287710q26.13IntergenicCT0.80Risk1.06 (1.04–1.08)4.11 × 10−8FGFR2[43]
rs3516656911p13IntergenicCT0.09 Protective0.89 (0.86–0.93)2.54 × 10−8WT1[42]
rs1103179611p13IntronAG0.31 Protective0.93 (0.91–0.94)2.47 × 10−15WT1-AS[42]
rs1074227711p13IntronCG0.33Risk1.48 (1.29–1.68)6.72 × 10−9WT1[43]
rs488677815q24.1IntronAC0.47 Risk1.05 (1.03–1.07)4.12 × 10−8LOXL1[42]
rs23592921q21.3IntronCG0.39 Protective0.93 (0.92–0.95)2.01 × 10−12ADAMTS5, ADAMTS1[42]
rs223647921q22.3IntronAG0.59 Risk2.232.80 × 10−7COL18A1[44]
Abbreviations: SNP: single nucleotide polymorphism; POP: pelvic organ prolapse; OR: odds ratio; EAF: effect allele frequency; CI: confidence interval; UTR: untranslated region; * significant results were indicated by p < 0.05 in candidate gene association studies and by p < 5 × 10−8 in genome-wide association studies (p < 1 × 10−7 was seen as genome-wide significant in reference [44]); ^ for intergenic variants, nearby gene(s) is(are) reported.
Table 2. Summary of significant SNPs associated with hormone metabolism in candidate gene association studies and genome-wide association studies.
Table 2. Summary of significant SNPs associated with hormone metabolism in candidate gene association studies and genome-wide association studies.
Candidate Gene Association Studies
ComponentSNP IDAnnotationChange in NucleotideCandidate GeneEffect DirectionSignificant Association with POP *Race/EthnicitySample Size (POP/Control)Reference
Estrogen
receptor
rs17847075/rs2077647ExonicT>CESR1Riskgenotype TC, OR 2.7East Asian (minority/non-minority Chinese)88 vs. 108[33]
rs2234693IntronicT>CESR1Riskgenotype TC, OR 2.99East Asian (minority/non-minority Chinese)88 vs. 108[33]
rs2228480ExonicG>AESR1Riskgenotype GA, OR 2.05East Asian (Chinese)88 vs. 153[75]
Riskgenotype AA, OR 39.70
genotype GA, OR 19.20
Ashkenazi-Jewish origin33 vs. 33[65]
Progestogen receptorrs484389ExonicT>CPGRRiskgenotype TC, OR 4.77East Asian (Chinese)87 vs. 150[76]
Genome-Wide Association Studies
SNP IDBand RegionAnnotationEffect AlleleOther AlleleEAFEffect DirectionOR (95% CI)p ValueMapped Gene(s) ^Reference
rs38202821p36.12IntronTC0.17Protective0.85 (0.82–0.88)3.30 × 10−21WNT4[41,42]
rs7283976817p13.1ExonAG0.02Risk1.19 (1.12–1.26)4.66 × 10−9DVL2[42]
Abbreviations: SNP: single nucleotide polymorphism; POP: pelvic organ prolapse; OR: odds ratio; EAF: effect allele frequency; CI: confidence interval; UTR: untranslated region; * significant results were indicated by p < 0.05 in candidate gene association studies and by p < 5 × 10−8 in genome-wide association studies (p < 1 × 10−7 was seen as genome-wide significant in reference [44]); ^ for intergenic variants, nearby gene(s) is(are) reported.
Table 3. Summary of significant SNPs associated with OS and others in candidate gene association studies and genome-wide association studies.
Table 3. Summary of significant SNPs associated with OS and others in candidate gene association studies and genome-wide association studies.
Candidate Gene Association Studies
ComponentSNP IDAnnotationChange in NucleotideCandidate GeneEffect DirectionSignificant Association with POP *Race/EthnicitySample Size (POP/Control)Reference
OSrs1695ExonicA>GGSTP1Protectivegenotype AG+GG, OR 0.63
allele G, OR 0.60
East Asian (Korean)189 vs. 156[89]
OSrs1136410ExonicT>CPARP1Protectivegenotype CC, OR 0.46
allele C, OR 0.72
East Asian (Korean)185 vs. 155[90]
Genome-Wide Association Studies
SNP IDBand RegionAnnotationEffect AlleleOther AlleleEAFEffect DirectionOR (95% CI)p ValueMapped Gene(s) ^Reference
OS-related
rs10368198q24.22IntronCA0.31Risk4.033.57 × 10−21ZFAT[44]
rs181063620p13IntronCA0.57Risk2.326.06 × 10−8IDH3B[44]
rs226737222q13.1IntronGA0.61Protective0.93 (0.91–0.95)1.07 × 10−13MAFF[42]
The variants overlapped with other metabolic and cardiovascular health
rs1076263110q22.1IntronAG0.10 Protective0.92 (0.90–0.95)3.76 × 10−8ADK[42]
rs1231424312p13.2IntronTC0.54 Protective0.91 (0.90–0.93)3.66 × 10−9DUSP16[42]
rs7319735312q24.21IntergenicCT0.08 Risk1.12 (1.08–1.17)1.63 × 10−8TBX5[42]
rs124794312q24.21IntergenicAG0.12 Risk1.09 (1.06–1.12)1.68 × 10−21TBX5[41,42]
rs477951715q13.2IntronGC0.49 Risk1.07 (1.05–1.09)1.10 × 10−11KLF13[42]
Others
rs581701203q21.3IntergenicAT0.18 Risk1.08 (1.06–1.11)1.17 × 10−10SEC61A1[42]
rs2011949994q13.2IntergenicTC0.30 Protective0.89 (0.86–0.93)2.42 × 10−8EPHA5[42]
rs14553114q21.21IntronGA0.34 Risk2.587.65 × 10−12PAQR3, BMP2K, ANTXR2[44]
rs284032754q28.1IntergenicCG0.18 Risk1.12 (1.10–1.15)1.58 × 10−22FAT4[42]
rs76829924q28.1IntergenicTA0.21 Risk1.13 (1.10–1.16)4.50 × 10−16FAT4[41]
rs100137694q28.1IntergenicGA0.65 Risk1.07 (1.05–1.09)1.26 × 10−10FAT4[42]
rs2512175q23.3IntronGA0.61 Risk1.06 (1.05–1.08)4.22 × 10−11SLC12A2, FBN2[42]
rs726249767q32.13’UTRTC0.01 Protective0.84 (0.79–0.89)1.14 × 10−9IMPDH1[41,42]
rs14932028q13.2IntergenicGT0.52 Risk1.05 (1.03–1.07)3.56 × 10−8LACTB2[42]
rs4307949q22.2IntronTG0.13 Protective0.356.74 × 10−5AUH, NFIL3[44]
rs648416111p15.4IntronTG0.31 Risk1.06 (1.04–1.08)5.89 × 10−9SBF2, ADM[42]
rs494493611q13.4IntergenicCT0.72 Protective0.93 (0.91–0.95)7.13 × 10−12CHRDL2[42]
rs8027714 15q11.2Intergenic AG0.26 Risk9.045.65 × 10−43NPAP1[44]
rs1291555415q13.13’UTRAC0.32 Protective0.95 (0.93–0.96)1.06 × 10−8GREM1[42]
rs1232519216q21.1IntergenicTC0.18 Protective0.89 (0.87–0.91)1.14 × 10−21SALL1[41,42]
rs187400816q24.13’UTRCT0.77 Protective0.94 (0.92–0.96)5.77 × 10−9CRISPLD2[42]
Abbreviations: SNP: single nucleotide polymorphism; POP: pelvic organ prolapse; OR: odds ratio; EAF: effect allele frequency; CI: confidence interval; OS: oxidative stress; UTR: untranslated region; * significant results were indicated by p < 0.05 in candidate gene association studies and by p < 5 × 10−8 in genome-wide association studies (p < 1 × 10−7 was seen as genome-wide significant in reference [44]); ^ for intergenic variants, nearby gene(s) is(are) reported.
Table 4. Summary of POP-associated genes and their expression in different pelvic floor tissues.
Table 4. Summary of POP-associated genes and their expression in different pelvic floor tissues.
ComponentsTissuesGeneRNAProteinRace/EthnicitySample Size (POP/Control)Reference
& Methods# POP vs. ControlName& Methods# POP vs.
Control
ECM-related
Collagen
Cardinal ligament---Type III collagenIHCCaucasian33 vs. 25[26]
---Type III collagenIHC, WBEast Asian (Chinese)30 vs. 30[113]
Uterosacral ligament---Type III collagenIHCEuropean (German)25 vs. 16[45]
---Type III collagenIHCTurk22 vs. 23[114]
COL3A1qRT-PCRType III collagenIHCEast Asian (Chinese)22 vs. 34
COL3A1qRT-PCRND---Turk32 vs. 8[115]
COL3A1qRT-PCRNDType III collagenIHCNDEast Asian (Chinese)35 vs. 20[116]
COL3A1qRT-PCRType III collagenIHCEast Asian (Chinese)30 vs. 30[117]
Round ligamentCOL3A1qRT-PCRND---Turk32 vs. 8[115]
Para-urethral tissuesCOL3A1qRT-PCRNDType III collagenIHCNDEuropean (Sweden)15 vs. 14[118]
Vaginal wall---Type III collagenIFAmerican62 vs. 15[119]
---Type III collagenIHCEast Asian (Chinese)23 vs. 15[120]
COL3A1qRT-PCR---American47 vs. 7[121]
---Type III collagenWBAmerican17 vs. 5[122]
---Type III collagenIHCNDCaucasian13 vs. 13[123]
---Type III collagenIHC, IFEuropean (Italian)14 vs. 10[124]
COL3A1qRT-PCRType III collagenIHCEast Asian (Chinese)60 vs. 35[125]
---Type III collagenIHC, WBEuropean (Italian)20 vs. 10[126]
---Type III collagenIHC, WBEast Asian (Chinese)35 vs. 35[127]
---Type IV collagenIHCNDEast Asian (Chinese)23 vs. 15[120]
---Type V collagenIFNDAmerican62 vs. 15[119]
---Type V collagenIHCNDEast Asian (Chinese)23 vs. 15[120]
Elastic fibers
Cardinal ligament---ElastinIHCCaucasian33 vs. 25[26]
Uterosacral ligament---ElastinIHC, IFEuropean (German)59 vs. 30[128]
---ElastinIHCNDEast Asian (Chinese)30 vs. 30[58]
Vaginal wall---ElastinIHCEuropean (Belgian)15 vs. 0[129]
---ElastinIHCNDEast Asian (Chinese)23 vs. 15[120]
ELNqRT-PCRND---American47 vs. 7[121]
---ElastinIHCNDCaucasian13 vs. 13[123]
Uterosacral ligamentEFEMP1qRT-PCRNDFibulin-3IHCNDAmerican8 vs. 8[130]
Vaginal wallEFEMP1qRT-PCRNDFibulin-3IHCNDEast Asian (Korean)12 vs. 12[131]
Cardinal ligament---Fibulin-5IHCEast Asian (Chinese)53 vs. 25[132]
Uterosacral ligamentFBLN5qRT-PCR---American31 vs. 29[133]
FBLN5qRT-PCRFibulin-5WBEast Asian (Korean)30 vs. 30[134]
FBLN5qRT-PCRFibulin-5IHCAmerican8 vs. 8[130]
FBLN5--Fibulin-5IHCEast Asian (Chinese)30 vs. 30[58]
Para-urethral tissuesFBLN5qRT-PCRFibulin-5IHCNDEuropean (Sweden)15 vs. 14[118]
Vaginal wallFBLN5qRT-PCRFibulin-5IHCAmerican12 vs. 10[135]
FBLN5qRT-PCRND---Caucasian15 vs. 11[136]
Lysyl oxidase
Cardinal ligament---Lysyl oxidase homolog 1IHCEast Asian (Chinese)53 vs. 25[132]
Uterosacral ligamentLOXL1qRT-PCRLysyl oxidase homolog 1WBEast Asian (Korean)30 vs. 30[134]
---Lysyl oxidase homolog 1IHCEast Asian (Chinese)30 vs. 30[58]
Vaginal wallLOXL1qRT-PCRLysyl oxidase homolog 1IHC, WBNDCaucasian15 vs. 11[136]
Vaginal wallLOXL4qRT-PCRND---Caucasian15 vs. 11[136]
Glycoprotein
Vaginal wall---LamininIHCNDEast Asian (Chinese)23 vs. 15[120]
Extracellular proteases
Uterosacral ligament---Interstitial collagenaseIHCEuropean (Croatian)40 vs. 40[137]
---Interstitial collagenaseIHCEuropean (Croatian)46 vs. 49[138]
---Interstitial collagenaseIHCIsraeli20 vs. 20[139]
---Interstitial collagenaseIHCTurk42 vs. 49[140]
MMP1qRT-PCRNDInterstitial collagenaseIHCNDEast Asian (Chinese)35 vs. 20[116]
Round ligament---Interstitial collagenaseIHCTurk42 vs. 49[140]
Vaginal wall---Interstitial collagenaseIHCIsraeli20 vs. 20[139]
MMP1qRT-PCRNDInterstitial collagenaseIHC, IBNDCaucasian17 vs. 19[141]
MMP1qRT-PCRInterstitial collagenaseIHCEast Asian (Chinese)72 vs. 72[142]
MMP1qRT-PCRInterstitial collagenaseIHCEast Asian (Chinese)60 vs. 35[125]
Cardinal ligament---Matrix metalloproteinase-9IHC, WBEast Asian (Chinese)30 vs. 30[113]
Uterosacral ligament---Matrix metalloproteinase-9IHCIsraeli20 vs. 20[139]
MMP9qRT-PCRMatrix metalloproteinase-9ELISAEast Asian (Korean)35 vs. 39[143]
---Matrix metalloproteinase-9IHCNDAmerican21 vs. 19[144]
MMP9qRT-PCRMatrix metalloproteinase-9IHCEast Asian (Chinese)35 vs. 20[116]
Vaginal wall---Matrix metalloproteinase-9IFAmerican62 vs. 15[119]
---Matrix metalloproteinase-9IHCIsraeli20 vs. 20[139]
MMP9qRT-PCRNDMatrix metalloproteinase-9IHC, IBNDCaucasian17 vs. 19[141]
Vaginal wallMMP10qRT-PCRND---American47 vs. 7[121]
Uterosacral ligamentTIMP2qRT-PCRMetalloproteinase inhibitor 2IHCEast Asian (Chinese)19 vs. 9[145]
TIMP2qRT-PCRMetalloproteinase inhibitor 2IHCNDEast Asian (Chinese)35 vs. 20[116]
Vaginal wallTIMP2qRT-PCRMetalloproteinase inhibitor 2IHC, IBNDCaucasian17 vs. 19[141]
Cervix tissueTIMP2qRT-PCRNDMetalloproteinase inhibitor 2IHCNDEast Asian (Chinese)19 vs. 9[145]
Hormone metabolism-related
Estrogen receptor
Uterosacral ligament---ER-αWBEast Asian (Korean)20 vs. 24[146]
ESR1qRT-PCRER-αIHCCaucasian13 vs. 13[147]
ESR1qRT-PCR---Turk32 vs. 8[115]
ESR1qRT-PCRER-αIHCEast Asian (Chinese)35 vs. 20[116]
Round ligamentESR1qRT-PCR---Turk32 vs. 8[115]
Progesterone receptor
Uterosacral ligament---Progesterone receptorWBEast Asian (Korean)20 vs. 24[146]
PGRqRT-PCRNDProgesterone receptorIHCNDCaucasian13 vs. 13[147]
Abbreviations: POP: pelvic organ prolapse; ECM: extracellular matrix; # “-” indicates that it was not analyzed in original studies; “↑” indicates that there was an increase in POP group; “↓” indicates that there was an decrease in POP group; “ND” indicates that there was no difference between two groups in original studies. & Methods: IHC: immunohistochemistry; qRT-PCR: real-time reverse transcription polymerase chain reaction; WB: Western blotting; IB: immunoblotting; IF: immunofluorescence; ELISA: enzyme-linked immunosorbent assay.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jiang, W.; Cheung, R.Y.K.; Chung, C.Y.; Chan, S.S.C.; Choy, K.W. Genetic Etiology in Pelvic Organ Prolapse: Role of Connective Tissue Homeostasis, Hormone Metabolism, and Oxidative Stress. Genes 2025, 16, 5. https://doi.org/10.3390/genes16010005

AMA Style

Jiang W, Cheung RYK, Chung CY, Chan SSC, Choy KW. Genetic Etiology in Pelvic Organ Prolapse: Role of Connective Tissue Homeostasis, Hormone Metabolism, and Oxidative Stress. Genes. 2025; 16(1):5. https://doi.org/10.3390/genes16010005

Chicago/Turabian Style

Jiang, Wenxuan, Rachel Yau Kar Cheung, Cheuk Yan Chung, Symphorosa Shing Chee Chan, and Kwong Wai Choy. 2025. "Genetic Etiology in Pelvic Organ Prolapse: Role of Connective Tissue Homeostasis, Hormone Metabolism, and Oxidative Stress" Genes 16, no. 1: 5. https://doi.org/10.3390/genes16010005

APA Style

Jiang, W., Cheung, R. Y. K., Chung, C. Y., Chan, S. S. C., & Choy, K. W. (2025). Genetic Etiology in Pelvic Organ Prolapse: Role of Connective Tissue Homeostasis, Hormone Metabolism, and Oxidative Stress. Genes, 16(1), 5. https://doi.org/10.3390/genes16010005

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop