Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Cervical microbiota dysbiosis associated with high-risk Human Papillomavirus infection

  • Natalia Zeber-Lubecka ,

    Roles Conceptualization, Formal analysis, Funding acquisition, Investigation, Project administration, Supervision, Visualization, Writing – original draft, Writing – review & editing

    natalia.zeber-lubecka@cmkp.edu.pl

    Affiliations Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland, Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Maria Kulecka,

    Roles Data curation, Formal analysis, Methodology, Software, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland, Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Michalina Dabrowska,

    Roles Investigation, Writing – review & editing

    Affiliation Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Katarzyna Baginska-Drabiuk,

    Roles Investigation, Writing – review & editing

    Affiliation Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Maria Glowienka-Stodolak,

    Roles Investigation, Writing – review & editing

    Affiliation Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Andrzej Nowakowski,

    Roles Resources, Writing – review & editing

    Affiliation Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Aneta Slabuszewska-Jozwiak,

    Roles Resources, Writing – review & editing

    Affiliation First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, Warsaw, Poland

  • Bożena Bednorz,

    Roles Resources, Writing – review & editing

    Affiliation Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Ilona Jędrzejewska,

    Roles Resources, Writing – review & editing

    Affiliation Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Magdalena Piasecka,

    Roles Resources, Writing – review & editing

    Affiliation Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Jolanta Pawelec,

    Roles Resources, Writing – review & editing

    Affiliation Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Elzbieta Wojciechowska-Lampka,

    Roles Funding acquisition, Writing – review & editing

    Affiliation Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

  • Jerzy Ostrowski

    Roles Funding acquisition, Project administration, Supervision, Writing – original draft, Writing – review & editing

    Affiliations Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland, Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland

Abstract

High-risk Human Papillomavirus (HR-HPV) genotypes, specifically HPV16 and HPV18, pose a significant risk for the development of cervical intraepithelial neoplasia and cervical cancer. In the multifaceted cervical microenvironment, consisting of immune cells and diverse microbiota, Lactobacillus emerges as a pivotal factor, wielding significant influence in both stabilizing and disrupting the microbiome of the reproductive tract. To analyze the distinction between the cervical microbiota and Lactobacillus-dominant/non-dominant status of HR-HPV and non-infected healthy women, sixty-nine cervical swab samples were analyzed, included 44 with HR-HPV infection and healthy controls. All samples were recruited from Human Papillomavirus-based cervical cancer screening program and subjected to 16s rRNA sequencing analysis. Alpha and beta diversity analyses reveal no significant differences in the cervical microbiota of HR-HPV-infected women, including 16 and 18 HPV genotypes, and those with squamous intraepithelial lesion (SIL), compared to a control group. In this study we identified significantly lower abundance of Lactobacillus mucosae in women with HR-HPV infection compared to the control group. Furthermore, changes in bacterial diversity were noted in Lactobacillus non-dominant (LND) samples compared to Lactobacillus-dominant (LD) in both HR-HPV-infected and control groups. LND samples in HR-HPV-infected women exhibited a cervical dysbiotic state, characterized by Lactobacillus deficiency. In turn, the LD HR-HPV group showed an overrepresentation of Lactobacillus helveticus. In summary, our study highlighted the distinctive roles of L. mucosae and L. helveticus in HR-HPV infections, signaling a need for further research to demonstrate potential clinical implications of cervical microbiota dysbiosis.

Introduction

Persistent infections with high-risk Human Papillomavirus (HR-HPV) genotypes–HPV16 or HPV18 –constitute a major risk factor for cervical intraepithelial neoplasia (CIN) and cervical cancer (CC) [1]. HR-HPV infections were found in 99.7% of CC case studies [1]. The estimated lifetime risk of infection with any type of HPV is about 80%, while the risk of developing CC is only 0.6% [2]. In fact, about 90% of HR-HPV infections are transient and resolve spontaneously [3]. In recent years, preventing HPV infection has emerged as a major health priority [4]. Significant strides have been made in primary prevention, largely due to the introduction of HPV vaccines, which have demonstrated high efficacy in preventing infection. Studies suggest that transitioning from first-generation HPV vaccines to nonavalent vaccines could potentially enhance prevention rates for both low- and high-grade squamous intraepithelial lesions (L- and HSILs) in nearly 90% of cases [5].

The microbiota found across diverse areas of the female genital tract may display both similarities and notable distinctions [68]. The vaginal, cervical and cervico-vaginal microbiota collectively refer to the diverse array of microorganisms, encompassing bacteria, viruses, and fungi, residing in the vagina and on the cervix [9]. Several factors: hormonal fluctuations, sexual activity, hygiene practices, as well as contraceptive and antibiotic usage influence the composition of genital tract microbiota [1012]. In contrast to the intestinal microflora, which is characterized by a large diversity of microorganisms, the genital tract of healthy women mainly contain one or several species of lactic acid bacilli, such as Lactobacillus crispatus, L. gasseri, L. iners, L. jensenii and L. acidophilus [13]. In turn, bacterial vaginosis is associated with the elimination of Lactobacillus spp. that become replaced by a diverse mix of anaerobic bacteria, such as Anaerococcus, Atopobium, Dialister, Fusobacterium, Gardnerella, Gemella, Prevotella, Megasphaera, Parvimonas, Peptoniphilus, and Peptostreptococcus [14]. Lactobacillus spp. play a range of important roles in the homeostasis of the cervical and vaginal microenvironment, such as anti-inflammatory effects that improve barrier function. Lactobacillus species produce lactic acid, which acidifies the local microenvironment to pH <4.5. protecting against the development of inflammation [15]. Moreover, metabolites produced by Lactobacillus spp. stimulate the production of antimicrobial peptides and anti-inflammatory cytokines [13].

The microbial communities found in the vagina and cervico-vaginal environment have been categorized into five major Community State Types (CSTs). However, the current CSTs classification offers only a partial comprehension of how the microbiota relates to cervico-vaginal conditions in women, largely due to limitations in bacterial identification technologies [16]. These categories are characterized by the predominance of specific Lactobacillus species. CST-I is primarily dominated by L. crispatus, CST-II by L. gasseri, CST-III by L. iners, and CST-V by L. jensenii. In contrast, CST-IV exhibits a more diverse microbial composition [17]. France et al. introduced the VALENCIA (Vaginal Community State Type Nearest Centroid) classifier tool, which enables consistent assignment of CSTs within the vaginal microbiota of reproductive-age women [18].

In women diagnosed with HR-HPV infection, long-term alterations in the cervico-vaginal microbiota composition positively correlated with microbial diversity at two time points, six months apart [16]. Women who initially exhibited a high abundance of L. iners tended to maintain a more stable microbiome composition in subsequent visits compared to those with communities depleted of Lactobacillus at the outset. Certain species including L. acidophilus and Megasphaera genomosp type 1 were linked to changes in CSTs between visits [16]. Persistent HPV infection and CIN development were associated with higher abundance of Gardnerella, Prevotella, and Megasphaera in cervical and cervico-vaginal microbiota [1921]. Moreover, some research revealed a trend of co-infection between HPV and syphilis, caused by Treponema pallidum, in certain populations [22]. In vitro experiments have shed light on potential mechanisms by which microbiota influence HPV infections, including the disruption of the cervical epithelial barrier through the regulation of adherence junction proteins, modulation of cervical immune responses, and alteration of miRNA expression [19]. However, SILs and progression to precancerous interepithelial neoplasia and CC are the clinical outcomes of HPV infection, the role of cervical dysbiosis in tumorigenesis remains unclear [14, 2329].

Considering all the mentioned dependencies, in this study we conducted a comparison between a group of women infected with HR-HPV and non-infected healthy women to identify differences in cervical microbiota profiles according to the HPV infection and Lactobacillus non-dominant status using 16S rRNA gene amplicon sequencing.

Materials and methods

Patients and controls recruitment

The study was conducted according to the guidelines of the 1964 Declaration of Helsinki and approved by the Bioethics Committee at the Maria Sklodowska-Curie National Research Institute of Oncology in Warsaw Review Board (No 42/2021). All participants provided informed consent to participate. Between 2nd August 2021 and 30th September 2022 enrolled HPV-infected and healthy women were recruited under the Human Papillomavirus-based cervical cancer screening program in the Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology. The study cohort included Polish Caucasian women who had provided written informed consent before being included in the study. Inclusion criteria for the study encompassed women older than 18 years, who were sexually active and unvaccinated for HPV. None of the participants were undergoing hormone replacement therapy. Exclusion criteria consisted of pregnancy and lactation, a history of resection surgery, hysterectomy, or pelvic radiotherapy, HIV infection, vaginal douching, or antibiotic therapy.

Sample collection procedure

Papanicolaou test (Pap test) examination and HPV DNA testing.

During the cervical screening visit, the patients underwent Pap test examination first. Brushes with exfoliated cervical cells preserved in 20 ml SurePath® Preservative Solution were subjected to liquid-based cytology and testing for the presence of HPV DNA. HPV infection was assayed using the Cobas4800HPV test for 3 HR signals of HPV types 16, 18 and others (Roche Molecular Diagnostics, Pleasanton, CA). A microscopic screening for precancerous cervical lesions based on the Bethesda System which divides epithelial cell abnormalities into low- and high-grade squamous intraepithelial lesion (L/HSIL) categories. CIN 1 and CIN 2/3 were classified as LSIL and HSIL, respectively. Additionally, equivocal morphological features including atypical squamous cells (ASCs) were subdivided into two categories: ASC of Undetermined Significance (ASC-US) and ASC of HSIL (ASC-H).

Cervical swab samples collection, DNA extraction and 16S rRNA sequencing for microbiota analysis.

After Pap test examination, cervical swab samples were collected aseptically from each patient using 4N6FLOQSwabs™ (Thermo Fisher Scientific, MA, USA), transferred to a collection tube and stored at −80°C within 30 minutes of collection. Bacterial genomic DNA was extracted from 4N6FLOQSwabs™ (Thermo Fisher Scientific, MA, USA) samples using the PureLink™ Microbiome DNA Purification Kit, according to the manufacturer’s instructions. The quantity and purity of extracted DNA were measured with the Nanodrop ND-1000 spectrophotometer (Thermo Fisher Scientific, MA, USA). Cervical microbiota analysis was performed using the Ion Torrent technology, following the taxonomic identification of bacteria based on hypervariable fragments of the 16S rRNA gene sequencing (Ion 16S™ Metagenomics Kit; Thermo Fisher Scientific, MA, USA) as described previously [30].

Statistical and bioinformatic analysis.

Unmapped BAM files were converted to FASTQ using Picard’s SamToFastq. Additional steps of the analysis were performed using Mothur software version 1.43 [31]. FASTQ files were converted to the FASTA format. We only included sequences that were 200–300 bases in length, with an average base quality of 20 in a sliding window of 50 bases, and a maximum homopolymer length of 10. Chimeric sequences were identified with the vsearch algorithm using the default parameters, with internal sequence collection as the reference database [32]. Chimeric sequences were removed and the remaining 16S rRNA sequences were classified using the Wang method and the SILVA [33] bacterial 16S rRNA database for reference (release 138). The bootstrap cut-off was 80%. Alpha diversity analysis was performed using the Shannon and Chao index as an indicator. Principal coordinate analysis (PCoA) was performed using the Bray-Curtis index as a distance measure. The ANOSIM was used to test the significance of clustering patterns. The differential abundance of taxa was assessed using the LinDA with the default parameters [34]. The Mann-Whitney U-test was utilized to assess variations in diversity indices between control and HR-HPV samples, while the Wilcoxon paired test was employed to identify statistically significant differences within paired patient samples. Adjusted p-values below 0.05, after controlling for false discovery rate (FDR), were deemed as indicative of significance. Lactobacillus species was determined by reassigning reads to the Greengenes database, version 13_8 [35]. The patients were defined as Lactobacillus-dominant if the bacteria abundance was higher than 70% [36].

Results

Patients and samples, HPV genotype testing and histopathologic diagnoses

Ultimately, 46 women diagnosed with a HR-HPV infection (median age 40 years, range 25–59) were enrolled in the study. The control group comprised 27 healthy women (median age 42 years, range 27–58) who tested negative for HPV and exhibited normal Pap tests. Nevertheless, the quantity and quality of DNA isolated from cervical swab samples enabled the construction of libraries from 44 HR-HPV-positive and 25 HPV-negative samples. Therefore, in the final analysis, a total of 69 samples were utilized for the study of cervical microbiota.

The characteristics of HR-HPV-infected women according to the Bethesda System is presented in Table 1. In this group, 13 infected women were diagnosed with SIL (and 3 of them were HSIL). Three HR-HPV infected women with SIL also presented ASC-US features. ASC-US and ASC-H were detected in 4 and 1 women without SIL, respectively. No-intraepithelial lesions (NILM) without SIL or ASC findings were shown in another 26 HR-HPV- infected women. The Pap test was normal in all HPV-negative women. Two HPV-infected women used intrauterine contraceptive devices (IUD).

thumbnail
Table 1. Characteristics of total HR-HPV-infected women based on Pap tests results according to the Bethesda system.

https://doi.org/10.1371/journal.pone.0302270.t001

Cervical microbiota diversity

Of 894 taxa (131 of which were found in over 0.01% of reads), 454 taxa were present in both groups, and 376 and 64 were detected only in HR-HPV patients and healthy controls, respectively (Fig 1). In total, we detected an average of 100 taxa per sample and 178517 reads. At the OTU level, microbial richness, and alpha and beta diversity were estimated using Chao, Shannon and PCoA indices, respectively. Alpha diversity analysis showed no statistically significant differences in the cervical microbiota of women infected with HPV compared to the control group (Fig 2A and 2B). A similar result was obtained after clustering the HPV types, HR-HPV 16 and 18, compared to the group of patients infected with other HR-HPV (Fig 2C and 2D). Also, PCoA showed no statistical differences in the beta diversity of cervical microbiota between HR-HPV and controls (Fig 3A). A comparable result was obtained comparing 16 and 18 HR-HPV patients to other HR-HPV and controls (Fig 3B).

thumbnail
Fig 1. Krona charts of the taxon abundance with a mean >1% of the total reads found in the HR-HPV (A) and control (B) samples.

https://doi.org/10.1371/journal.pone.0302270.g001

thumbnail
Fig 2.

Shannon index (A, C), and Chao index (B, D) of the cervical microbiota between the HR-HPV, 16 and 18 or other HR-HPV genotypes and control group.

https://doi.org/10.1371/journal.pone.0302270.g002

thumbnail
Fig 3.

Principal coordinates analysis based on the Bray–Curtis index for HR-HPV (A), 16 and 18 or other HR-HPV genotypes (B) and control group.

https://doi.org/10.1371/journal.pone.0302270.g003

At the genus level, only unclassified Comamonadaceae differentiated HR-HPV-infected patients from controls (padj <0.05) whose lower amounts were found in HR-HPV samples. The abundance of other species, unclassified Rhizobiaceae and Aminobacter, tended to be statistically significant and was also lower in HR-HPV women (Table 2).

thumbnail
Table 2. Bacteria at the genus level differentiating HR-HPV and control samples.

https://doi.org/10.1371/journal.pone.0302270.t002

Next, we compared the samples from HR-HPV-positive women with or without SIL and those from the healthy controls. Again, both alpha (Fig 4A and 4B) and beta diversity (Fig 4C) of cervical microbiota did not differ between the studied groups. Due to the limited size of the patient group with Pap-test lesions, statistically significant results were not attainable. The only difference related to the significantly lower abundance of unclassified Comamonadaceae genus was demonstrated in non-SIL patient samples compared to controls.

thumbnail
Fig 4.

Shannon index (A), Chao index (B) and Principal coordinates analysis based on the Bray–Curtis index (C) of the cervical microbiota between the HR-HPV with and without squamous intraepithelial lesion (SIL) and control group.

https://doi.org/10.1371/journal.pone.0302270.g004

Lactobacilli findings

Lactobacillus species were found in all 25 HPV-negative and in 41 out of 44 HR-HPV-positive women (Fig 5A and 5B, respectively). Of these, Lactobacillus dominated (with the bacteria abundance higher than 70%) in the cervical microbiota of 17, from total 25 HPV-negative and 27, from total 44 HR-HPV-positive women. The distribution of predominant Lactobacillus species was presented in Table 3.

thumbnail
Fig 5.

Heat-map diagram of the cervical microbiota composition at genus level for HR-HPV (A) and control (B) groups.

https://doi.org/10.1371/journal.pone.0302270.g005

thumbnail
Table 3. Lactobacillus distribution in Lactobacillus-dominant HR-HPV-positive and HPV-negative samples (lactobacilli threshold >70%).

https://doi.org/10.1371/journal.pone.0302270.t003

Seventeen HR-HPV-positive samples and 8 HPV-negative samples were classified as Lactobacillus non-dominant (LND). Several pathogenic bacteria were detected in HPV-positive and LND samples. They were classified as CST type IV, including Streptococcus, Prevotella, Gardnerella, Streptococcus, Atopobium, Megasphaera and Anaerococcus genera (Table 4). In HPV-negative and LND samples, we identified Gardnerella, Prevotella and Streptococcus in 4, 2 and 1 samples, respectively.

thumbnail
Table 4. Bacterial distribution in Lactobacillus non-dominant HR-HPV-positive and control samples.

https://doi.org/10.1371/journal.pone.0302270.t004

When the cervical microbiota of HR-HPV-infected women was compared to those of control group, one species of lactobacilli, L. mucosae was found to be less represented in the HR-HPV group (padj 0.02; FC -2.37). In contrast, no differentiating lactobacilli were found in the cervical microbiota of women with SIL compared to both the control and HR-HPV non-SIL groups.

Cervical microbiota dominated vs non-dominated by Lactobacillus

In both LND groups of women infected with HR-HPV and controls we observed a higher bacterial diversity compared to the LD (Lactobacillus-dominant) group (Shannon index padj 1.1e-09 and 0.00025, respectively) (Fig 6A and 6D). Furthermore, the groups also differed as regards the beta diversity level (ANOSIM p = 0.0001, R = 0.703; ANOSIM p = 0.0001, R = 0.789) (Fig 6C and 6F) but did not differ as regards bacterial richness (Fig 6B and 6E). The abundance of 23 taxa significantly differentiated LD from LND samples in women infected with HR-HPV (Table 5). Among these, 5 and 18 taxa were under- and overrepresented, respectively in LND HR-HPV samples.

thumbnail
Fig 6.

Shannon index (A, D), Chao index (B, E) and Principal coordinates analysis based on the Bray–Curtis index (C, F) in comparisons of Lactobacillus-dominant (LD) vs. Lactobacillus non-dominant (LND) of HR-HPV and control groups.

https://doi.org/10.1371/journal.pone.0302270.g006

thumbnail
Table 5. Taxa distinguishing LD from LND samples in women infected with HR-HPV.

https://doi.org/10.1371/journal.pone.0302270.t005

Notably, a similar analysis conducted in the control group revealed only one statistically significant taxon (Bifidobacteriaceae unclassified, padj 0.046) distinguishing LD from LND. Even though Lactobacillus helveticus was overrepresented in LD HR-HPV group (FC 2.53; padj 0.029) as compared to LND HR-HPV group, no distinguishing lactobacilli were found by the same comparisons in the control group.

Discussion

The cervicovaginal mucosa relies on several key defense mechanisms, including antimicrobial peptides and microbiota primarily composed of lactobacilli. Any disruptions to these protective mechanisms may lead to physical and chemical changes that harm the cervical epithelium and vaginal mucosa [37]. Specifically, a reduction in lactobacilli, which are responsible for producing lactic acid, can promote abnormal bacterial growth and reduce the presence of protective flora, thereby weakening the body’s natural defense system against viral infections [38].

Several studies have explored the potential influence of HPV infection on the cervico-vaginal microbiota and vice versa [3941]. However, exact mechanisms and causal relationships are still being elucidated. Dysbiosis, or imbalance, in the cervico-vaginal microbiota has been associated with HPV persistence, which is a key factor in the development of cervical precancerous lesions and cancer [42]. Certain bacteria stimulate immune cells and modulate inflammation, potentially affecting the clearance or persistence of HPV [43]. While HPV vaccination has reduced the burden of CC, non-vaccine-preventable HPV genotypes still pose a risk [44, 45]. Crucially, not every woman infected with HR-HPV develop CC, leading to investigations into potential protective factors. The hypotheses imply that beneficial bacteria, including L. acidophilus in the cervico-vaginal microbiota, could contribute to safeguarding against CC development among HR-HPV-infected women [46]. Detailed microbiome profiling in a Dutch cervical cancer screening program found that women with typical cervical smears and a higher abundance of L. acidophilus were associated with a lower risk of HSIL, highlighting the role of this bacteria in cervico-vaginal microbiota dynamics and continuity [46].

The primary objective of this study was to investigate the variances in cervical microbiota profiles between two groups: women infected with HR-HPV and non-infected healthy women. We aimed to discern distinctions in cervical microbiota characteristics concerning HR-HPV infection and the prevalence of non-dominant Lactobacillus species.

The analysis of alpha and beta diversity revealed no significant differences in the cervical microbiota of women infected with HPV, including HR-HPV types 16 and 18, as well as the SIL, compared to the control group. A limitation of this study is related to the fact that only 10 LSIL and 3 HSIL samples were included. Unclassified Comamonadaceae were the differentiating bacteria of the HR-HPV groups and controls, SIL and non-SIL. It seems that the occurrence of these differentiating bacteria in the cervix was related to HPV infection but not to SIL. A lower abundance of Comamonadaceae in HR-HPV-infected women was also observed in two studies conducted by Chen et al. [47, 48].

Liu et al. showed that the Shannon index of vaginal microbiota diversity was higher in women with HPV infection, signifying a greater number of bacteria compared to HPV-negative women [49]. Moreover, a lower Simpson’s index in HPV-positive women indicated the complexity of the vaginal microflora enhanced by HPV infection but reduced under stable healthy vaginal conditions. However, the bacterial richness did not show a significant relationship with HPV infection. As shown in previous studies, significant differences in cervical microbiota diversity were only observed in HSIL [41, 50]. Guo et al. reported a significantly higher alpha diversity in the HPV+HSIL group compared to the HPV+LSIL group [41]. Nevertheless, no significant differences were found between HPV+LSIL and HPV+NoSIL, nor between HPV+NoSIL and HPV-negative groups. Additionally, PCoA revealed only slight variations in microbial samples among the HPV-negative, HPV+NoSIL, HPV+LSIL, and HPV+HSIL groups [41]. Lin et al. demonstrated no significant changes in Shannon diversity for CIN in the vagina and cervix, even though the number of observed OTUs in the cervical specimens was slightly higher for CIN than in healthy controls [50]. However, microbial diversity increased significantly in cervical specimens compared to their corresponding vaginal specimens in CIN. Furthermore, when examining different subtypes of CIN, the Shannon diversity in the cervical microbiota significantly increased in the CIN 1 subtype. Additionally, Comamonas (Comamonadaceae), Rhizobium (Rhizobiaceae), and Pseudomonas (Pseudomonadaceae) were identified as bacteria strongly linked with CIN in the cervix [50].

The most remarkable changes in bacterial diversity affect women who have developed CC. In a case-control analysis involving healthy individuals and patients diagnosed with either CIN 2/3 or invasive CC, microbial richness was notably elevated in that group compared to the controls [51]. The increase was associated with a higher count of OTUs. Our previous study [30] revealed that cervical microbiota diversity measured by the Shannon index was significantly greater in pre-treatment CC women in comparison with control samples. However, we did not observe significant differences in species richness. These results reveal significant alterations in the cervical microbiota of CC patients relative to that in healthy controls.

It is the first study which revealed that among various lactobacilli studied, only L. mucosae exhibited a significantly lower presence in the group of women with HR-HPV infection compared to the control group. In contrast, our analysis did not reveal any significant differences in the composition of lactobacilli in the cervical microbiota of women with SIL when compared to both the control group and the HR-HPV non-SIL group. L. mucosae inhabits the digestive systems of both humans and animals, and it is also found in the human vaginal environment. L. mucosae produces exopolysaccharides (EPS), which exhibit anti-inflammatory effects [52]. This bacterium is known for its ability to neutralize toxins and exhibit antimicrobial activity, effectively controlling harmful substances including Zen toxin and inhibiting various Gram-positive and Gram-negative pathogens, including Escherichia coli, Salmonella typhimurium and Staphylococcus [53, 54]. As of now, the presence of Lactobacillus mucosae in the cervix has not been described or documented in scientific literature. Moreover, previous research indicated that Lactobacillus mucosae is not commonly found in the vaginal microbiota [55]. Das Purkayastha et al. found a relative dominance of vaginal L. mucosae compared to L. crispatus and L. jensenii among both non-pregnant and pregnant women of Northeast India [56]. The authors suggested that L. mucosae might have a competitive advantage over other bacteria in terms of binding to receptors and utilizing nutrients within the vaginal epithelial cells [56]. L. mucosae was documented to produce the Lam29 protein which allowed it to effectively outcompete pathogens, safeguarding the intestinal mucosa [57]. Similarly, in the human vaginal environment, the epithelial layer serves as a protective barrier that pathogens seek to breach, while Lactobacillus aims to protect it by adhering and outcompeting the intruders. Since a lower abundance of L. mucosae may be a result of HPV infection, we hypothesize that a loss or decrease in the abundance of L. mucosae may lead to deficiencies in its protective function among HPV-infected women.

In both HR-HPV-infected and control groups, changes in bacterial diversity were observed in LND samples compared to LD. Such a result is consistent with previous studies [5860]. Additionally, LND samples in HR-HPV-infected women were classified as CST IV, characterized as a dysbiotic state which includes Lactobacillus deficiency and consists of a diverse group of microorganisms, including both strictly obligate and facultative anaerobes [17, 61, 62]. Conversely, the LD HR-HPV group showed an overrepresentation of L. helveticus, which produces hydrogen peroxide, impedes the attachment of pathogens, and decreases the viability of bacteria associated with bacterial vaginosis [63]. During both the follicular and luteal phases, L. helveticus emerged as the predominant species [64]. Kim et al. confirmed the potential of L. helveticus HY7801 as a beneficial probiotic for the physiological vaginal state [65]. In vivo experiments showed that HY7801 reduced the presence of Gardnerella vaginalis and pro-inflammatory cytokines in BV-induced mice, improving histological changes of vaginal tissues. Importantly, it was found that HY7801 could alleviate bacterial vaginosis by inhibiting the virulence factor genes of Gardnerella vaginalis related to cell adhesion and biofilm formation, along with antibacterial activity against G. vaginalis [65]. Moreover, Scibior-Bentkowska et al. showed that an increased abundance of L. helveticus, L. suntoryeus, and L. vaginalis in women with persistent HR-HPV infection was associated with a potentially protective effect against the development of CIN 3 [66].

Conclusions

Our study provided insights into the association between the cervical microbiota, HR-HPV infections, and Lactobacillus non-dominant status. We showed for the first time that only L. mucosae exhibited a significantly lower abundance in the group of women with HR-HPV infection in comparison with the controls. Alternatively, further research is needed before potential clinical implications of cervical microbiota dysbiosis could be generalized.

References

  1. 1. Graham SV. The human papillomavirus replication cycle, and its links to cancer progression: a comprehensive review. Clinical Science 2017;131:2201–21. pmid:28798073
  2. 2. Kombe Kombe AJ, Li B, Zahid A, Mengist HM, Bounda G-A, Zhou Y, et al. Epidemiology and Burden of Human Papillomavirus and Related Diseases, Molecular Pathogenesis, and Vaccine Evaluation. Front Public Health 2021;8:552028. pmid:33553082
  3. 3. Huber J, Mueller A, Sailer M, Regidor P-A. Human papillomavirus persistence or clearance after infection in reproductive age. What is the status? Review of the literature and new data of a vaginal gel containing silicate dioxide, citric acid, and selenite. Womens Health (Lond Engl) 2021;17:174550652110207. pmid:34096424
  4. 4. Calagna G, Maranto M, Paola C, Capra G, Perino A, Chiantera V, et al. ‘Secondary prevention’ against female HPV infection: literature review of the role of carrageenan. Expert Review of Anti-Infective Therapy 2020;18:865–74. pmid:32552158
  5. 5. Capra G, Giovannelli L, Matranga D, Bellavia C, Guarneri MF, Fasciana T, et al. Potential impact of a nonavalent HPV vaccine on HPV related low-and high-grade cervical intraepithelial lesions: A referral hospital-based study in Sicily. Human Vaccines & Immunotherapeutics 2017;13:1839–43. pmid:28594305
  6. 6. Gholiof M, Adamson-De Luca E, Wessels JM. The female reproductive tract microbiotas, inflammation, and gynecological conditions. Front Reprod Health 2022;4:963752. pmid:36303679
  7. 7. Kumar L, Dwivedi M, Jain N, Shete P, Solanki S, Gupta R, et al. The Female Reproductive Tract Microbiota: Friends and Foe. Life 2023;13:1313. pmid:37374096
  8. 8. Plesniarski A, Siddik AB, Su R-C. The Microbiome as a Key Regulator of Female Genital Tract Barrier Function. Front Cell Infect Microbiol 2021;11:790627. pmid:34976864
  9. 9. Głowienka-Stodolak M, Bagińska-Drabiuk K, Szubert S, Hennig EE, Horala A, Dąbrowska M, et al. Human Papillomavirus Infections and the Role Played by Cervical and Cervico-Vaginal Microbiota-Evidence from Next-Generation Sequencing Studies. Cancers (Basel) 2024;16:399. pmid:38254888
  10. 10. Moosa Y, Kwon D, de Oliveira T, Wong EB. Determinants of Vaginal Microbiota Composition. Front Cell Infect Microbiol 2020;10. pmid:32984081
  11. 11. Kroon SJ, Ravel J, Huston WM. Cervicovaginal microbiota, women’s health, and reproductive outcomes. Fertil Steril 2018;110:327–36. pmid:30098679
  12. 12. Cascardi E, Cazzato G, Daniele A, Silvestris E, Cormio G, Di Vagno G, et al. Association between Cervical Microbiota and HPV: Could This Be the Key to Complete Cervical Cancer Eradication? Biology (Basel) 2022;11:1114. pmid:35892970
  13. 13. Łaniewski P, Ilhan ZE, Herbst-Kralovetz MM. The microbiome and gynaecological cancer development, prevention and therapy. Nat Rev Urol 2020;17:232–50. pmid:32071434
  14. 14. Mitra A, MacIntyre DA, Lee YS, Smith A, Marchesi JR, Lehne B, et al. Cervical intraepithelial neoplasia disease progression is associated with increased vaginal microbiome diversity. Sci Rep 2015;5:16865. pmid:26574055
  15. 15. Martin DH, Marrazzo JM. The Vaginal Microbiome: Current Understanding and Future Directions. J Infect Dis 2016;214:S36–41. pmid:27449871
  16. 16. Molina MA, Andralojc KM, Huynen MA, Leenders WPJ, Melchers WJG. In-depth insights into cervicovaginal microbial communities and hrHPV infections using high-resolution microbiome profiling. NPJ Biofilms Microbiomes 2022;8:75. pmid:36171433
  17. 17. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SSK, McCulle SL, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A 2011;108 Suppl 1:4680–7. pmid:20534435
  18. 18. France MT, Ma B, Gajer P, Brown S, Humphrys MS, Holm JB, et al. VALENCIA: a nearest centroid classification method for vaginal microbial communities based on composition. Microbiome 2020;8:166. pmid:33228810
  19. 19. Kawahara R, Fujii T, Kukimoto I, Nomura H, Kawasaki R, Nishio E, et al. Changes to the cervicovaginal microbiota and cervical cytokine profile following surgery for cervical intraepithelial neoplasia. Sci Rep 2021;11:2156. pmid:33495564
  20. 20. Oh HY, Kim B-S, Seo S-S, Kong J-S, Lee J-K, Park S-Y, et al. The association of uterine cervical microbiota with an increased risk for cervical intraepithelial neoplasia in Korea. Clin Microbiol Infect 2015;21:674.e1–9. pmid:25752224
  21. 21. Zhai Q, Zhang W, Zhang Z, Fu Y, Li Y, Wang X, et al. Characteristics of the Cervicovaginal Microenvironment in Childbearing-Age Women with Different Degrees of Cervical Lesions and HR-HPV Positivity. Pol J Microbiol 2021;70:489–500. pmid:34970317
  22. 22. Fasciana T, Capra G, Di Carlo P, Calà C, Vella M, Pistone G, et al. Socio-Demographic Characteristics and Sexual Behavioral Factors of Patients with Sexually Transmitted Infections Attending a Hospital in Southern Italy. IJERPH 2021;18:4722. pmid:33925258
  23. 23. Reimers LL, Mehta SD, Massad LS, Burk RD, Xie X, Ravel J, et al. The Cervicovaginal Microbiota and Its Associations With Human Papillomavirus Detection in HIV-Infected and HIV-Uninfected Women. J Infect Dis 2016;214:1361–9. pmid:27521363
  24. 24. Di Paola M, Sani C, Clemente AM, Iossa A, Perissi E, Castronovo G, et al. Characterization of cervico-vaginal microbiota in women developing persistent high-risk Human Papillomavirus infection. Sci Rep 2017;7:10200. pmid:28860468
  25. 25. Brusselaers N, Shrestha S, Van De Wijgert J, Verstraelen H. Vaginal dysbiosis and the risk of human papillomavirus and cervical cancer: systematic review and meta-analysis. American Journal of Obstetrics and Gynecology 2019;221:9–18.e8. pmid:30550767
  26. 26. Berggrund M, Gustavsson I, Aarnio R, Lindberg JH, Sanner K, Wikström I, et al. Temporal changes in the vaginal microbiota in self-samples and its association with persistent HPV16 infection and CIN2+. Virol J 2020;17:147. pmid:33028395
  27. 27. Dareng EO, Ma B, Adebamowo SN, Famooto A, Ravel J, Pharoah PP, et al. Vaginal microbiota diversity and paucity of Lactobacillus species are associated with persistent hrHPV infection in HIV negative but not in HIV positive women. Sci Rep 2020;10:19095. pmid:33154533
  28. 28. Tamarelle J, Thiébaut ACM, De Barbeyrac B, Bébéar C, Ravel J, Delarocque-Astagneau E. The vaginal microbiota and its association with human papillomavirus, Chlamydia trachomatis, Neisseria gonorrhoeae and Mycoplasma genitalium infections: a systematic review and meta-analysis. Clinical Microbiology and Infection 2019;25:35–47. pmid:29729331
  29. 29. Usyk M, Zolnik CP, Castle PE, Porras C, Herrero R, Gradissimo A, et al. Cervicovaginal microbiome and natural history of HPV in a longitudinal study. PLoS Pathog 2020;16:e1008376. https://doi.org/10.1371/journal.ppat.1008376.
  30. 30. Zeber-Lubecka N, Kulecka M, Lindner B, Krynicki R, Paziewska A, Nowakowski A, et al. Increased diversity of a cervical microbiome associates with cervical cancer. Front Oncol 2022;12:1005537. pmid:36249017
  31. 31. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing mothur: Open-Source, Platform-Independent, Community-Supported Software for Describing and Comparing Microbial Communities. Appl Environ Microbiol 2009;75:7537–41. pmid:19801464
  32. 32. Rognes T, Flouri T, Nichols B, Quince C, Mahé F. VSEARCH: a versatile open source tool for metagenomics. PeerJ 2016;4:e2584. pmid:27781170
  33. 33. Silva CJ, Caparica AA, Plascak JA. Wang-Landau Monte Carlo simulation of the Blume-Capel model. Phys Rev E 2006;73:036702. pmid:16605693
  34. 34. Zhou H, He K, Chen J, Zhang X. LinDA: linear models for differential abundance analysis of microbiome compositional data. Genome Biol 2022;23:95. pmid:35421994
  35. 35. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a Chimera-Checked 16S rRNA Gene Database and Workbench Compatible with ARB. Appl Environ Microbiol 2006;72:5069–72. pmid:16820507
  36. 36. Miller EA, Beasley DE, Dunn RR, Archie EA. Lactobacilli Dominance and Vaginal pH: Why Is the Human Vaginal Microbiome Unique? Front Microbiol 2016;7. pmid:28008325
  37. 37. Lacroix G, Gouyer V, Gottrand F, Desseyn J-L. The Cervicovaginal Mucus Barrier. International Journal of Molecular Sciences 2020;21:8266. pmid:33158227
  38. 38. Mizutani T, Ishizaka A, Koga M, Tsutsumi T, Yotsuyanagi H. Role of Microbiota in Viral Infections and Pathological Progression. Viruses 2022;14:950. pmid:35632692
  39. 39. Vargas-Robles D, Romaguera J, Alvarado-Velez I, Tosado-Rodríguez E, Dominicci-Maura A, Sanchez M, et al. The cervical microbiota of Hispanics living in Puerto Rico is nonoptimal regardless of HPV status. mSystems 2023;8:e00357–23. pmid:37534938
  40. 40. Li C, Zhang Z, Yang Y, Liao H. Changes in the cervicovaginal microbiota composition of HPV16-infected patients after clinical treatment. Cancer Medicine 2022;11:5037–49. pmid:35569127
  41. 41. Guo C, Dai W, Zhou Q, Gui L, Cai H, Wu D, et al. Cervicovaginal microbiota significantly changed for HPV-positive women with high-grade squamous intraepithelial lesion. Frontiers in Cellular and Infection Microbiology 2022;12. pmid:35992171
  42. 42. Zeng M, Li X, Jiao X, Cai X, Yao F, Xu S, et al. Roles of vaginal flora in human papillomavirus infection, virus persistence and clearance. Front Cell Infect Microbiol 2023;12:1036869. pmid:36683675
  43. 43. Castanheira CP, Sallas ML, Nunes RAL, Lorenzi NPC, Termini L. Microbiome and Cervical Cancer. Pathobiology 2020;88:187–97. pmid:33227782
  44. 44. Buttà M, Serra N, Panzarella V, Fasciana TMA, Campisi G, Capra G. Orogenital Human Papillomavirus Infection and Vaccines: A Survey of High- and Low-Risk Genotypes Not Included in Vaccines. Vaccines 2023;11:1466. pmid:37766142
  45. 45. Choi S, Ismail A, Pappas-Gogos G, Boussios S. HPV and Cervical Cancer: A Review of Epidemiology and Screening Uptake in the UK. Pathogens 2023;12:298. pmid:36839570
  46. 46. Andralojc KM, Molina MA, Qiu M, Spruijtenburg B, Rasing M, Pater B, et al. Novel high-resolution targeted sequencing of the cervicovaginal microbiome. BMC Biol 2021;19:267. pmid:34915863
  47. 47. Chen Y, Hong Z, Wang W, Gu L, Gao H, Qiu L, et al. Association between the vaginal microbiome and high-risk human papillomavirus infection in pregnant Chinese women. BMC Infect Dis 2019;19:677. pmid:31370796
  48. 48. Chen Y, Qiu X, Wang W, Li D, Wu A, Hong Z, et al. Human papillomavirus infection and cervical intraepithelial neoplasia progression are associated with increased vaginal microbiome diversity in a Chinese cohort. BMC Infect Dis 2020;20:629. pmid:32842982
  49. 49. Liu S, Li Y, Song Y, Wu X, Baloch Z, Xia X. The diversity of vaginal microbiome in women infected with single HPV and multiple genotype HPV infections in China. Front Cell Infect Microbiol 2022;12:642074. pmid:36601309
  50. 50. Lin S, Zhang B, Lin Y, Lin Y, Zuo X. Dysbiosis of Cervical and Vaginal Microbiota Associated With Cervical Intraepithelial Neoplasia. Frontiers in Cellular and Infection Microbiology 2022;12. pmid:35237529
  51. 51. Tango CN, Seo S-S, Kwon M, Lee D-O, Chang HK, Kim MK. Taxonomic and Functional Differences in Cervical Microbiome Associated with Cervical Cancer Development. Sci Rep 2020;10:9720. pmid:32546712
  52. 52. Jurášková D, Ribeiro SC, Silva CCG. Exopolysaccharides Produced by Lactic Acid Bacteria: From Biosynthesis to Health-Promoting Properties. Foods 2022;11:156. pmid:35053888
  53. 53. Valeriano VD, Parungao-Balolong MM, Kang D-K. In vitro evaluation of the mucin-adhesion ability and probiotic potential of Lactobacillus mucosae LM1. J Appl Microbiol 2014;117:485–97. https://doi.org/10.1111/jam.12539.
  54. 54. Soriano AP, Mamuad LL, Kim S-H, Choi YJ, Jeong CD, Bae GS, et al. Effect of Lactobacillus mucosae on In vitro Rumen Fermentation Characteristics of Dried Brewers Grain, Methane Production and Bacterial Diversity. Asian Australas J Anim Sci 2014;27:1562–70. https://doi.org/10.5713/ajas.2014.14517.
  55. 55. Madhivanan P, Raphael E, Rumphs A, Krupp K, Ravi K, Srinivas V, et al. Characterization of culturable vaginal Lactobacillus species among women with and without bacterial vaginosis from the United States and India: a cross-sectional study. Journal of Medical Microbiology 2014;63:931–5. pmid:24836413
  56. 56. Das Purkayastha S, Bhattacharya MK, Prasad HK, Upadhyaya H, Lala SD, Pal K, et al. Contrasting diversity of vaginal lactobacilli among the females of Northeast India. BMC Microbiol 2019;19:198. pmid:31455211
  57. 57. Watanabe M, Kinoshita H, Huang I-N, Eguchi K, Tsurumi T, Kawai Y, et al. An Adhesin-Like Protein, Lam29, from Lactobacillus mucosae ME-340 Binds to Histone H3 and Blood Group Antigens in Human Colonic Mucus. Bioscience, Biotechnology, and Biochemistry 2012;76:1655–60. https://doi.org/10.1271/bbb.120150.
  58. 58. Tuddenham S, Gajer P, Burke AE, Murphy C, Klein SL, Stennett CA, et al. Lactobacillus-dominance and rapid stabilization of vaginal microbiota in combined oral contraceptive pill users examined through a longitudinal cohort study with frequent vaginal sampling over two years. eBioMedicine 2023;87:104407. pmid:36529102
  59. 59. Tortelli BA, Lewis AL, Fay JC. The structure and diversity of strain-level variation in vaginal bacteria. Microbial Genomics 2021;7. pmid:33656436
  60. 60. Chee WJY, Chew SY, Than LTL. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb Cell Fact 2020;19:203. pmid:33160356
  61. 61. Shannon B, Yi TJ, Perusini S, Gajer P, Ma B, Humphrys MS, et al. Association of HPV infection and clearance with cervicovaginal immunology and the vaginal microbiota. Mucosal Immunology 2017;10:1310–9. pmid:28120845
  62. 62. Fettweis JM, Brooks JP, Serrano MG, Sheth NU, Girerd PH, Edwards DJ, et al. Differences in vaginal microbiome in African American women versus women of European ancestry. Microbiology 2014;160:2272–82. pmid:25073854
  63. 63. Atassi F, Brassart D, Grob P, Graf F, Servin AL. In vitro antibacterial activity of Lactobacillus helveticus strain KS300 against diarrhoeagenic, uropathogenic and vaginosis-associated bacteria. J Appl Microbiol 2006;101:647–54. pmid:16907815
  64. 64. Cheng L, Gao Y, Xia Q, Wang H, Xie X, Liu Y, et al. Reproductive tract microbiota of women in childbearing age shifts upon gynecological infections and menstrual cycle. BMC Microbiol 2021;21:252. pmid:34548031
  65. 65. Kim JY, Moon EC, Kim J-Y, Kim HJ, Heo K, Shim J-J, et al. Lactobacillus helveticus HY7801 ameliorates bacterial vaginosis by inhibiting biofilm formation and epithelial cell adhesion of Gardnerella vaginalis. Food Sci Biotechnol 2023;32:507–15. pmid:36911333
  66. 66. Scibior-Bentkowska D, Banila C, Nedjai B. 2022-RA-1195-ESGO Longitudinal study of vaginal microbiome pre- and post-treatment identifies biomarkers for cervical intraepithelial neoplasia 3 (CIN3). Translational research/biomarkers, BMJ Publishing Group Ltd; 2022, p. A413.2–A414. https://doi.org/10.1136/ijgc-2022-ESGO.886.