Bacterial vaginosis and health-associated bacteria modulate the immunometabolic landscape in 3D model of human cervix

Paweł Łaniewski, Melissa M Herbst-Kralovetz, Paweł Łaniewski, Melissa M Herbst-Kralovetz

Abstract

Bacterial vaginosis (BV) is an enigmatic polymicrobial condition characterized by a depletion of health-associated Lactobacillus and an overgrowth of anaerobes. Importantly, BV is linked to adverse gynecologic and obstetric outcomes: an increased risk of sexually transmitted infections, preterm birth, and cancer. We hypothesized that members of the cervicovaginal microbiota distinctly contribute to immunometabolic changes in the human cervix, leading to these sequelae. Our 3D epithelial cell model that recapitulates the human cervical epithelium was infected with clinical isolates of cervicovaginal bacteria, alone or as a polymicrobial community. We used Lactobacillus crispatus as a representative health-associated commensal and four common BV-associated species: Gardnerella vaginalis, Prevotella bivia, Atopobium vaginae, and Sneathia amnii. The immunometabolic profiles of these microenvironments were analyzed using multiplex immunoassays and untargeted global metabolomics. A. vaginae and S. amnii exhibited the highest proinflammatory potential through induction of cytokines, iNOS, and oxidative stress-associated compounds. G. vaginalis, P. bivia, and S. amnii distinctly altered physicochemical barrier-related proteins and metabolites (mucins, sialic acid, polyamines), whereas L. crispatus produced an antimicrobial compound, phenyllactic acid. Alterations to the immunometabolic landscape correlate with symptoms and hallmarks of BV and connected BV with adverse women's health outcomes. Overall, this study demonstrated that 3D cervical epithelial cell colonized with cervicovaginal microbiota faithfully reproduce the immunometabolic microenvironment previously observed in clinical studies and can successfully be used as a robust tool to evaluate host responses to commensal and pathogenic bacteria in the female reproductive tract.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Cervicovaginal Lactobacillus and BVAB effectively…
Fig. 1. Cervicovaginal Lactobacillus and BVAB effectively colonize the surface and crevices of human 3D cervical epithelial cell model.
Scanning electron micrographs of human 3D cervical models infected with L. crispatus JV-V01 (a), A. vaginae CCUG 38953 (b), S. amnii Sn35 (c), G. vaginalis JCP8151B (d), P. bivia VPI 6822 (e), or a polymicrobial community (consisting of equal parts of four BVAB: G. vaginalis, P. bivia, S. amnii, and A. vaginae) (orange) (f). Bacterial cells had expected shapes (i.e., rod-shaped bacilli for L. crispatus or small coccobacilli for BVAB species). Bacterial cells attached to surfaces of epithelial cells and formed clusters, which interacted with multiple epithelial cells. Scale bars on micrographs show 5 μm in distance. Bacteria were pseudocolored using the Affinity Designer software: cyan blue for L. crispatus, red for A. vaginae, yellow for S. amnii, green for G. vaginalis, purple for P. bivia, and orange for the polymicrobial cocktail. These colors are also used in the following figures to indicate particular bacterial infections.
Fig. 2. BVAB species distinctively modulate host…
Fig. 2. BVAB species distinctively modulate host defense responses in human 3D cervical models relative to Lactobacillus and PBS controls.
A. vaginae, S. amnii, and polymicrobial community exerted the greatest proinflammatory potentials, whereas G. vaginalis and P. bivia mostly altered the epithelial barrier targets. S. amnii also induced proteins related to cellular stress and angiogenesis. Human 3D cervical models were colonized with L. crispatus type strain (1) and L. crispatus vaginal isolate (2) or infected with single BVAB species and a polymicrobial community (consisting of four tested BVAB) for 24 h. The heatmaps depict relative levels of cytokines/chemokines (a), protein targets related to physicochemical barrier (b), and growth factors and cellular stress-related proteins (c) evaluated in the cell culture supernatants. Bacterial infections were grouped using hierarchical clustering. The data were mean centered and scaled and variance was scaled for each target. Clustering was based on Euclidean distance and average linkage. Black dots indicate significant changes (p < 0.05) in protein levels when compared to both Lactobacillus and uninfected (PBS) controls. Gray dots indicate targets significantly different from PBS only, whereas white dots indicate targets significantly different from Lactobacillus only. Statistical differences between the mean levels of protein targets among the groups were determined using ANOVA with Tukey’s adjustment for multiple comparisons.
Fig. 3. Human 3D cervical models infected…
Fig. 3. Human 3D cervical models infected with A. vaginae, S. amnii, and a polymicrobial community exert more similar metabolic profiles and cluster separately from G. vaginalis, P. bivia, and L. crispatus-colonized models or uninfected control.
Human 3D cervical models were colonized with L. crispatus type strain (1) and L. crispatus vaginal isolate (2) or infected with single BVAB species and a polymicrobial community (consisting of four tested BVAB) for 24 h. Global metabolomic analysis was performed on collected cell culture supernatants. a Total number of significantly (p < 0.05) altered metabolites following bacterial infections when compared to uninfected controls. The superpathways are indicated by colored bars. The polymicrobial infection altered the greatest number of metabolites, which mostly belonged to amino acid and nucleotide superpathways. b Number of unique and shared metabolites among the monomicrobial and polymicrobial BVAB infections visualized by the Venn diagram. Only five metabolites were shared by all the tested BVAB, indicating unique metabolic profiles of BVAB species. c Hierarchical clustering of all detected metabolites based on Euclidean distance and average linkage. The analysis revealed three separate clusters. Cell culture medium control clustered separately from all other groups. L. crispatus, G. vaginalis, and P. bivia clustered together with uninfected controls, whereas A. vaginae and S. amnii clustered with the polymicrobial infection. Statistical differences were determined using Welch’s two-sample t test.
Fig. 4. Metabolites in 3D cell culture…
Fig. 4. Metabolites in 3D cell culture supernatants highly predict monomicrobial and polymicrobial infection with BVAB or Lactobacillus.
Metabolome data were used to predict bacterial infections using Random Forest. The analysis revealed that the most predictive metabolites belong to amino acid and nucleotide superpathways, which were altered in species-specific manner. The analysis highlighted excellent predictive accuracy (93.75%) of infection, compared to random 12.5%. a Twenty most predictive metabolites are depicted and ranked by relative importance score. The superpathways are indicated by colored dots. b The heatmap shows fold changes of the most predictive metabolites among bacterial infections compared to uninfected controls. Only the fold changes that were significant (p < 0.05; q < 0.01) are displayed. Gold- and purple-shaded squares indicate metabolite enrichment or depletion, respectively. Statistical differences were determined using Welch’s two-sample t test with FDR correction. c The confusion matrix illustrates the proportion of times each sample receives the correct classification. Among the 36 tested samples, only 2 were confused (1 from L. crispatus-colonized models and 1 from P. bivia-infected models).
Fig. 5. Cervicovaginal Lactobacillus crispatus specifically alters…
Fig. 5. Cervicovaginal Lactobacillus crispatus specifically alters carbohydrate and amino acids pathways in a human 3D cervical model.
L. crispatus depletes glucose and alters energy metabolism, induce phenyllactate and N-acetylated amino acid production, and histidine degradation when compared to uninfected control and monomicrobial and polymicrobial BVAB infections. a The heatmap shows fold changes of the metabolites among bacterial infections compared to uninfected controls. Only the fold changes that were significant (p < 0.05) are displayed. Gold- and purple-shaded squares indicate metabolite enrichment or depletion, respectively. be Key metabolites altered by L. crispatus in human 3D cervical cell model relate to energy metabolism (b), synthesis of phenyllactate synthesis (c), N-acetylated amino acids (d), and histidine degradation pathway (e). The relative intensities of metabolites are shown as floating bar graphs. The boxes represent the median and interquartile range and whiskers range from minimum to maximum values. A plus sign (+) on plots indicates mean values. Schematics of glucose catabolism (b) and phenyllactate synthesis pathways (c) are also depicted. Colored circles indicate enriched or depleted metabolites within the pathway. Significant differences between mean intensities of metabolites among infections were determined using Welch’s two-sample t test with FDR correction. p and q values are indicated by ^(p < 0.05, q > 0.01), *(p < 0.05, q < 0.01), **(p < 0.01, q < 0.01), ***(p < 0.001, q < 0.01), ****(p < 0.0001, q < 0.01).
Fig. 6. BVAB induce production of polyamines…
Fig. 6. BVAB induce production of polyamines in a species-specific manner in a human 3D cervical model.
P. bivia and S. amnii contribute to agmatine, spermidine, and N(1)-acetylspermine production, whereas A. vaginae does not induce production of bioamines similarly to L. crispatus. a The heatmap shows fold changes of the metabolites related to the polyamine pathway among bacterial infections compared to uninfected controls. Only the fold changes that were significant (p < 0.05) are displayed. Gold- and purple-shaded squares indicate metabolite enrichment or depletion, respectively. b A schematic of polyamine metabolism pathways. Colored circles indicate enriched or depleted metabolites following monoinfection with specific BVAB or infection with the polymicrobial cocktail. c Floating bar graphs shows relative intensity of polyamines in 3D cell culture supernatants following bacterial colonization. The relative intensities of polyamines and related metabolites are shown as floating bar graphs. The boxes represent the median and interquartile range and whiskers range from minimum to maximum values. A plus sign (+) on plots indicates mean values. Significant differences between mean intensities of metabolites among infections were determined using Welch’s two-sample t test with FDR correction. p and q values are indicated by ^(p < 0.05, q > 0.01), *(p < 0.05, q < 0.01), **(p < 0.01, q < 0.01), ***(p < 0.001, q < 0.01).
Fig. 7. BVAB species distinctively contribute to…
Fig. 7. BVAB species distinctively contribute to inflammation and physicochemical attributes of epithelial barrier in a human 3D cervical model.
A. vaginae and S. amnii impact arginine and citrulline metabolism, which leads proinflammatory signaling via nitric oxide production (a). S. amnii also induce production of oxidative stress-related metabolites (b). S. amnii induce pipecolate associated with the presence of “clue cells” (c). P. bivia can contribute to elevated cervicovaginal pH levels via asparagine degradation and ammonia production (d). BVAB species also can impact physicochemical attributes of epithelial barrier (mucin degradation and collagen remodeling) (e, f). a A schematic of arginine/citrulline metabolism pathways. Colored circles indicate enriched or depleted metabolites following monoinfection with specific BVAB or infection with the polymicrobial cocktail. af Floating bar graphs shows relative intensity of polyamines in 3D cell culture supernatants following bacterial colonization. The relative intensities of metabolites are shown as floating bar graphs. The boxes represent the median and interquartile range and whiskers range from minimum to maximum values. A plus sign (+) on plots indicates mean values. Significant differences between mean intensities of metabolites among infections were corrected for multiple comparisons. p and q values are indicated by ^(p < 0.05, q > 0.01), *(p < 0.05, q < 0.01), **(p < 0.01, q < 0.01), ***(p < 0.001, q < 0.01), ****(p < 0.0001, q < 0.01).
Fig. 8. Summary of immunometabolic contributions of…
Fig. 8. Summary of immunometabolic contributions of Lactobacillus and BVAB species in the cervical microenvironment.
L. crispatus reinforces protective microenvironment via antimicrobial metabolites, including PLA. G. vaginalis and P. bivia alter physiochemical barrier-related proteins and metabolites (mucins, sialic acid, polyamines). A. vaginae and S. amnii induce robust proinflammatory and pro-oncogenic responses through induction of cytokines, iNOS, and oxidative stress. Polymicrobial infection leads to the most robust cervical immunometabolic activity. These mechanistic insights on immunometabolic landscape provide better understanding of BV pathogenesis and connect BV with adverse women’s health outcomes.

References

    1. Peebles K, Velloza J, Balkus JE, McClelland RS, Barnabas RV. High global burden and costs of bacterial vaginosis: a systematic review and meta-analysis. Sex. Transm. Dis. 2019;46:304–311.
    1. Koumans EH, et al. The prevalence of bacterial vaginosis in the United States, 2001-2004; associations with symptoms, sexual behaviors, and reproductive health. Sex. Transm. Dis. 2007;34:864–869.
    1. Amsel R, et al. Nonspecific vaginitis. Diagnostic criteria and microbial and epidemiologic associations. Am. J. Med. 1983;74:14–22.
    1. Muzny CA, Kardas P. A narrative review of current challenges in the diagnosis and management of bacterial vaginosis. Sex. Transm. Dis. 2020;47:441–446.
    1. Onderdonk AB, Delaney ML, Fichorova RN. The human microbiome during bacterial vaginosis. Clin. Microbiol. Rev. 2016;29:223–238.
    1. Ravel J, et al. Vaginal microbiome of reproductive-age women. Proc. Natl Acad. Sci. USA. 2011;108(Suppl. 1):4680–4687.
    1. Antonio MA, Hawes SE, Hillier SL. The identification of vaginal Lactobacillus species and the demographic and microbiologic characteristics of women colonized by these species. J. Infect. Dis. 1999;180:1950–1956.
    1. Martin DH, Marrazzo JM. The vaginal microbiome: current understanding and future directions. J. Infect. Dis. 2016;214(Suppl. 1):S36–S41.
    1. Younes JA, et al. Women and their microbes: the unexpected friendship. Trends Microbiol. 2018;26:16–32.
    1. Srinivasan S, et al. Bacterial communities in women with bacterial vaginosis: high resolution phylogenetic analyses reveal relationships of microbiota to clinical criteria. PLoS ONE. 2012;7:e37818.
    1. Marrazzo JM. A persistent(ly) enigmatic ecological mystery: bacterial vaginosis. J. Infect. Dis. 2006;193:1475–1477.
    1. Haggerty CL, et al. Bacterial vaginosis and anaerobic bacteria are associated with endometritis. Clin. Infect. Dis. 2004;39:990–995.
    1. Watts DH, Krohn MA, Hillier SL, Eschenbach DA. Bacterial vaginosis as a risk factor for post-cesarean endometritis. Obstet. Gynecol. 1990;75:52–58.
    1. Taylor BD, Darville T, Haggerty CL. Does bacterial vaginosis cause pelvic inflammatory disease? Sex. Transm. Dis. 2013;40:117–122.
    1. van Oostrum N, De Sutter P, Meys J, Verstraelen H. Risks associated with bacterial vaginosis in infertility patients: a systematic review and meta-analysis. Hum. Reprod. 2013;28:1809–1815.
    1. Wilson JD, Ralph SG, Rutherford AJ. Rates of bacterial vaginosis in women undergoing in vitro fertilisation for different types of infertility. BJOG. 2002;109:714–717.
    1. Schwebke JR, Weiss HL. Interrelationships of bacterial vaginosis and cervical inflammation. Sex. Transm. Dis. 2002;29:59–64.
    1. Hillier SL, et al. Association between bacterial vaginosis and preterm delivery of a low-birth-weight infant. The Vaginal Infections and Prematurity Study Group. N. Engl. J. Med. 1995;333:1737–1742.
    1. Klebanoff MA, Brotman RM. Treatment of bacterial vaginosis to prevent preterm birth. Lancet. 2018;392:2141–2142.
    1. Fettweis JM, et al. The vaginal microbiome and preterm birth. Nat. Med. 2019;25:1012–1021.
    1. Elovitz MA, et al. Cervicovaginal microbiota and local immune response modulate the risk of spontaneous preterm delivery. Nat. Commun. 2019;10:1305.
    1. Brown RG, et al. Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med. 2018;16:9.
    1. Nelson DB, et al. Early pregnancy changes in bacterial vaginosis-associated bacteria and preterm delivery. Paediatr. Perinat. Epidemiol. 2014;28:88–96.
    1. Doyle RM, et al. Bacterial communities found in placental tissues are associated with severe chorioamnionitis and adverse birth outcomes. PLoS ONE. 2017;12:e0180167.
    1. Lannon SMR, et al. Parallel detection of lactobacillus and bacterial vaginosis-associated bacterial DNA in the chorioamnion and vagina of pregnant women at term. J. Matern. Fetal Neonatal Med. 2019;32:2702–2710.
    1. Han YW, Shen T, Chung P, Buhimschi IA, Buhimschi CS. Uncultivated bacteria as etiologic agents of intra-amniotic inflammation leading to preterm birth. J. Clin. Microbiol. 2009;47:38–47.
    1. Vitorino P, et al. Sneathia amnii and maternal chorioamnionitis and stillbirth, Mozambique. Emerg. Infect. Dis. 2019;25:1614–1616.
    1. Cohen CR, et al. Bacterial vaginosis associated with increased risk of female-to-male HIV-1 transmission: a prospective cohort analysis among African couples. PLoS Med. 2012;9:e1001251.
    1. Martin HL, et al. Vaginal lactobacilli, microbial flora, and risk of human immunodeficiency virus type 1 and sexually transmitted disease acquisition. J. Infect. Dis. 1999;180:1863–1868.
    1. Cherpes TL, Meyn LA, Krohn MA, Lurie JG, Hillier SL. Association between acquisition of herpes simplex virus type 2 in women and bacterial vaginosis. Clin. Infect. Dis. 2003;37:319–325.
    1. Kaul R, et al. Prevalent herpes simplex virus type 2 infection is associated with altered vaginal flora and an increased susceptibility to multiple sexually transmitted infections. J. Infect. Dis. 2007;196:1692–1697.
    1. Chohan V, et al. A prospective study of risk factors for herpes simplex virus type 2 acquisition among high-risk HIV-1 seronegative women in Kenya. Sex. Transm. Infect. 2009;85:489–492.
    1. Watts DH, et al. Effects of bacterial vaginosis and other genital infections on the natural history of human papillomavirus infection in HIV-1-infected and high-risk HIV-1-uninfected women. J. Infect. Dis. 2005;191:1129–1139.
    1. Gillet E, et al. Bacterial vaginosis is associated with uterine cervical human papillomavirus infection: a meta-analysis. BMC Infect. Dis. 2011;11:10.
    1. Brotman RM, et al. Bacterial vaginosis assessed by gram stain and diminished colonization resistance to incident gonococcal, chlamydial, and trichomonal genital infection. J. Infect. Dis. 2010;202:1907–1915.
    1. Gallo MF, et al. Bacterial vaginosis, gonorrhea, and chlamydial infection among women attending a sexually transmitted disease clinic: a longitudinal analysis of possible causal links. Ann. Epidemiol. 2012;22:213–220.
    1. Wiesenfeld HC, Hillier SL, Krohn MA, Landers DV, Sweet RL. Bacterial vaginosis is a strong predictor of Neisseria gonorrhoeae and Chlamydia trachomatis infection. Clin. Infect. Dis. 2003;36:663–668.
    1. Lokken EM, et al. Association of recent bacterial vaginosis with acquisition of Mycoplasma genitalium. Am. J. Epidemiol. 2017;186:194–201.
    1. Nye MB, Harris AB, Pherson AJ, Cartwright CP. Prevalence of Mycoplasma genitalium infection in women with bacterial vaginosis. BMC Womens Health. 2020;20:62.
    1. Balkus JE, et al. Bacterial vaginosis and the risk of trichomonas vaginalis acquisition among HIV-1-negative women. Sex. Transm. Dis. 2014;41:123–128.
    1. Rathod SD, et al. Bacterial vaginosis and risk for Trichomonas vaginalis infection: a longitudinal analysis. Sex. Transm. Dis. 2011;38:882–886.
    1. Herbst-Kralovetz MM, Pyles RB, Ratner AJ, Sycuro LK, Mitchell C. New systems for studying intercellular interactions in bacterial vaginosis. J. Infect. Dis. 2016;214 Suppl 1:S6–S13.
    1. Radtke AL, Quayle AJ, Herbst-Kralovetz MM. Microbial products alter the expression of membrane-associated mucin and antimicrobial peptides in a three-dimensional human endocervical epithelial cell model. Biol. Reprod. 2012;87:132.
    1. Jackson R, Maarsingh JD, Herbst-Kralovetz MM, Van Doorslaer K. 3D oral and cervical tissue models for studying papillomavirus host-pathogen interactions. Curr. Protoc. Microbiol. 2020;59:e129.
    1. Barrila J, et al. Organotypic 3D cell culture models: using the rotating wall vessel to study host-pathogen interactions. Nat. Rev. Microbiol. 2010;8:791–801.
    1. Gardner JK, et al. Interleukin-36gamma is elevated in cervicovaginal epithelial cells in women with bacterial vaginosis and in vitro after infection with microbes associated with bacterial vaginosis. J. Infect. Dis. 2020;221:983–988.
    1. Salliss ME, Maarsingh JD, Garza C, Łaniewski P, Herbst-Kralovetz MM. Veillonellaceae family members uniquely alter the cervical metabolic microenvironment in a human three-dimensional epithelial model. NPJ Biofilms Microbiomes. 2021;7:57.
    1. McGowin CL, Radtke AL, Abraham K, Martin DH, Herbst-Kralovetz M. Mycoplasma genitalium infection activates cellular host defense and inflammation pathways in a 3-dimensional human endocervical epithelial cell model. J. Infect. Dis. 2013;207:1857–1868.
    1. Brygoo ER, Aladame N. [Study of a new strictly anaerobic species of the genus Eubacterium: Eubacterium crispatum n. sp.] Ann. Inst. Pasteur. 1953;84:640–641.
    1. Witkin SS, et al. Influence of vaginal bacteria and D- and L-lactic acid isomers on vaginal extracellular matrix metalloproteinase inducer: implications for protection against upper genital tract infections. MBio. 2013;4:e00460–00413.
    1. Lewis WG, Robinson LS, Gilbert NM, Perry JC, Lewis AL. Degradation, foraging, and depletion of mucus sialoglycans by the vagina-adapted Actinobacterium Gardnerella vaginalis. J. Biol. Chem. 2013;288:12067–12079.
    1. Holdeman LV, Johnson JL. Bacteroides disiens sp. nov. and Bacteroides bivius sp. nov. from human clinical infections. Int. J. Syst. Bacteriol. 1977;27:337–345.
    1. Rodriguez Jovita M, Collins MD, Sjoden B, Falsen E. Characterization of a novel Atopobium isolate from the human vagina: description of Atopobium vaginae sp. nov. Int. J. Syst. Bacteriol. 1999;49(Pt 4):1573–1576.
    1. Harwich MD, Jr., et al. Genomic sequence analysis and characterization of Sneathia amnii sp. nov. BMC Genomics. 2012;13(Suppl. 8):S4.
    1. Chetwin E, et al. Antimicrobial and inflammatory properties of South African clinical Lactobacillus isolates and vaginal probiotics. Sci. Rep. 2019;9:1917.
    1. Rose WA, 2nd, et al. Commensal bacteria modulate innate immune responses of vaginal epithelial cell multilayer cultures. PLoS ONE. 2012;7:e32728.
    1. Borgogna JC, et al. Biogenic amines increase the odds of bacterial vaginosis and affect the growth and lactic acid production by vaginal Lactobacillus spp. Appl. Environ. Microbiol. 2021 doi: 10.1128/AEM.03068-20.
    1. Ojala T, et al. Comparative genomics of Lactobacillus crispatus suggests novel mechanisms for the competitive exclusion of Gardnerella vaginalis. BMC Genomics. 2014;15:1070.
    1. Hung KJ, et al. Effect of commercial vaginal products on the growth of uropathogenic and commensal vaginal bacteria. Sci. Rep. 2020;10:7625.
    1. Abdelmaksoud AA, et al. Comparison of Lactobacillus crispatus isolates from Lactobacillus-dominated vaginal microbiomes with isolates from microbiomes containing bacterial vaginosis-associated bacteria. Microbiology. 2016;162:466–475.
    1. Gilbert NM, Lewis WG, Lewis AL. Clinical features of bacterial vaginosis in a murine model of vaginal infection with Gardnerella vaginalis. PLoS ONE. 2013;8:e59539.
    1. O’Brien VP, Joens MS, Lewis AL, Gilbert NM. Recurrent Escherichia coli urinary tract infection triggered by Gardnerella vaginalis bladder exposure in mice. J. Vis. Exp. 2020 doi: 10.3791/61967.
    1. Castro, J., Rosca, A. S., Muzny, C. A. & Cerca, N. Atopobium vaginae and Prevotella bivia are able to incorporate and influence gene expression in a pre-formed Gardnerella vaginalis biofilm. Pathogens10.3390/pathogens10020247 (2021).
    1. De Backer E, Dubreuil L, Brauman M, Acar J, Vaneechoutte M. In vitro activity of secnidazole against Atopobium vaginae, an anaerobic pathogen involved in bacterial vaginosis. Clin. Microbiol. Infect. 2010;16:470–472.
    1. Gentile GL, et al. Identification of a cytopathogenic toxin from Sneathia amnii. J. Bacteriol. 2020;202:e00162-00120.
    1. Muzny CA, et al. Identification of key bacteria involved in the induction of incident bacterial vaginosis: a prospective study. J. Infect. Dis. 2018;218:966–978.
    1. Linden SK, Sutton P, Karlsson NG, Korolik V, McGuckin MA. Mucins in the mucosal barrier to infection. Mucosal Immunol. 2008;1:183–197.
    1. Karantza V. Keratins in health and cancer: more than mere epithelial cell markers. Oncogene. 2011;30:127–138.
    1. Mitchell C, Marrazzo J. Bacterial vaginosis and the cervicovaginal immune response. Am. J. Reprod. Immunol. 2014;71:555–563.
    1. St John E, Mares D, Spear GT. Bacterial vaginosis and host immunity. Curr. HIV/AIDS Rep. 2007;4:22–28.
    1. Mu W, Yu S, Zhu L, Zhang T, Jiang B. Recent research on 3-phenyllactic acid, a broad-spectrum antimicrobial compound. Appl. Microbiol. Biotechnol. 2012;95:1155–1163.
    1. Srinivasan, S. et al. Metabolic signatures of bacterial vaginosis. MBio10.1128/mBio.00204-15 (2015).
    1. Vitali B, et al. Vaginal microbiome and metabolome highlight specific signatures of bacterial vaginosis. Eur. J. Clin. Microbiol. Infect. Dis. 2015;34:2367–2376.
    1. Genç MR, et al. Vaginal nitric oxide in pregnant women with bacterial vaginosis. Am. J. Reprod. Immunol. 2006;56:86–90.
    1. Holmes, K. K. Sexually Transmitted Diseases 4th edn (McGraw-Hill Medical, 2008).
    1. Mitchell CM, et al. Colonization of the upper genital tract by vaginal bacterial species in nonpregnant women. Am. J. Obstet. Gynecol. 2015;212:611.e611–611.e619.
    1. Quayle AJ. The innate and early immune response to pathogen challenge in the female genital tract and the pivotal role of epithelial cells. J. Reprod. Immunol. 2002;57:61–79.
    1. Bradshaw CS, et al. The association of Atopobium vaginae and Gardnerella vaginalis with bacterial vaginosis and recurrence after oral metronidazole therapy. J. Infect. Dis. 2006;194:828–836.
    1. Ferris MJ, et al. Association of Atopobium vaginae, a recently described metronidazole resistant anaerobe, with bacterial vaginosis. BMC Infect. Dis. 2004;4:5.
    1. Swidsinski A, et al. An adherent Gardnerella vaginalis biofilm persists on the vaginal epithelium after standard therapy with oral metronidazole. Am. J. Obstet. Gynecol. 2008;198:97 e91–96.
    1. Menard JP, et al. High vaginal concentrations of Atopobium vaginae and Gardnerella vaginalis in women undergoing preterm labor. Obstet. Gynecol. 2010;115:134–140.
    1. Baldwin EA, et al. Persistent microbial dysbiosis in preterm premature rupture of membranes from onset until delivery. PeerJ. 2015;3:e1398.
    1. Łaniewski P, et al. Linking cervicovaginal immune signatures, HPV and microbiota composition in cervical carcinogenesis in non-Hispanic and Hispanic women. Sci. Rep. 2018;8:7593.
    1. Mitra A, et al. Cervical intraepithelial neoplasia disease progression is associated with increased vaginal microbiome diversity. Sci. Rep. 2015;5:16865.
    1. Lee JE, et al. Association of the vaginal microbiota with human papillomavirus infection in a Korean twin cohort. PLoS ONE. 2013;8:e63514.
    1. Fichorova RN, Yamamoto HS, Delaney ML, Onderdonk AB, Doncel GF. Novel vaginal microflora colonization model providing new insight into microbicide mechanism of action. MBio. 2011;2:e00168-00111.
    1. Doerflinger SY, Throop AL, Herbst-Kralovetz MM. Bacteria in the vaginal microbiome alter the innate immune response and barrier properties of the human vaginal epithelia in a species-specific manner. J. Infect. Dis. 2014;209:1989–1999.
    1. Shannon B, et al. Distinct effects of the cervicovaginal microbiota and herpes simplex type 2 infection on female genital tract immunology. J. Infect. Dis. 2017;215:1366–1375.
    1. Masson L, et al. Inflammatory cytokine biomarkers of asymptomatic sexually transmitted infections and vaginal dysbiosis: a multicentre validation study. Sex. Transm. Infect. 2019;95:5–12.
    1. Dabee S, et al. Defining characteristics of genital health in South African adolescent girls and young women at high risk for HIV infection. PLoS ONE. 2019;14:e0213975.
    1. Lennard, K. et al. Microbial composition predicts genital tract inflammation and persistent bacterial vaginosis in South African adolescent females. Infect. Immun. 10.1128/IAI.00410-17 (2018).
    1. Spear GT, et al. Human alpha-amylase present in lower-genital-tract mucosal fluid processes glycogen to support vaginal colonization by Lactobacillus. J. Infect. Dis. 2014;210:1019–1028.
    1. Nunn, K. L. et al. Amylases in the human vagina. mSphere10.1128/mSphere.00943-20 (2020).
    1. van der Veer C, et al. Comparative genomics of human Lactobacillus crispatus isolates reveals genes for glycosylation and glycogen degradation: implications for in vivo dominance of the vaginal microbiota. Microbiome. 2019;7:49.
    1. Mirmonsef P, et al. Glycogen levels in undiluted genital fluid and their relationship to vaginal pH, estrogen, and progesterone. PLoS ONE. 2016;11:e0153553.
    1. Boskey ER, Cone RA, Whaley KJ, Moench TR. Origins of vaginal acidity: high D/L lactate ratio is consistent with bacteria being the primary source. Hum. Reprod. 2001;16:1809–1813.
    1. Kuchiiwa T, Nio-Kobayashi J, Takahashi-Iwanaga H, Yajima T, Iwanaga T. Cellular expression of monocarboxylate transporters in the female reproductive organ of mice: implications for the genital lactate shuttle. Histochem. Cell Biol. 2011;135:351–360.
    1. Lavermicocca P, et al. Purification and characterization of novel antifungal compounds from the sourdough Lactobacillus plantarum strain 21B. Appl. Environ. Microbiol. 2000;66:4084–4090.
    1. Hedegaard J, Brevet J, Roche J. Imidazole lactic acid: an intermediate in L-histidine degradation in Escherichia coli B. Biochem. Biophys. Res. Commun. 1966;25:335–339.
    1. Muzny CA, Łaniewski P, Schwebke JR, Herbst-Kralovetz MM. Host–vaginal microbiota interactions in the pathogenesis of bacterial vaginosis. Curr. Opin. Infect. Dis. 2020;33:59–65.
    1. Pybus V, Onderdonk AB. Evidence for a commensal, symbiotic relationship between Gardnerella vaginalis and Prevotella bivia involving ammonia: potential significance for bacterial vaginosis. J. Infect. Dis. 1997;175:406–413.
    1. Castro J, Machado D, Cerca N. Unveiling the role of Gardnerella vaginalis in polymicrobial bacterial vaginosis biofilms: the impact of other vaginal pathogens living as neighbors. ISME J. 2019;13:1306–1317.
    1. Chen KC, Forsyth PS, Buchanan TM, Holmes KK. Amine content of vaginal fluid from untreated and treated patients with nonspecific vaginitis. J. Clin. Investig. 1979;63:828–835.
    1. Chen KC, Amsel R, Eschenbach DA, Holmes KK. Biochemical diagnosis of vaginitis: determination of diamines in vaginal fluid. J. Infect. Dis. 1982;145:337–345.
    1. Nelson TM, et al. Vaginal biogenic amines: biomarkers of bacterial vaginosis or precursors to vaginal dysbiosis? Front. Physiol. 2015;6:253.
    1. Gong Z, et al. Arginine- and polyamine-induced lactic acid resistance in Neisseria gonorrhoeae. PLoS ONE. 2016;11:e0147637.
    1. Goytia M, Shafer WM. Polyamines can increase resistance of Neisseria gonorrhoeae to mediators of the innate human host defense. Infect. Immun. 2010;78:3187–3195.
    1. Ilhan ZE, Łaniewski P, Tonachio A, Herbst-Kralovetz MM. Members of Prevotella genus distinctively modulate innate immune and barrier functions in a human three-dimensional endometrial epithelial cell model. J. Infect. Dis. 2020 doi: 10.1093/infdis/jiaa324.
    1. Briselden AM, Moncla BJ, Stevens CE, Hillier SL. Sialidases (neuraminidases) in bacterial vaginosis and bacterial vaginosis-associated microflora. J. Clin. Microbiol. 1992;30:663–666.
    1. Hillier SL. Diagnostic microbiology of bacterial vaginosis. Am. J. Obstet. Gynecol. 1993;169:455–459.
    1. Cauci S, et al. Immunoglobulin A response against Gardnerella vaginalis hemolysin and sialidase activity in bacterial vaginosis. Am. J. Obstet. Gynecol. 1998;178:511–515.
    1. Agarwal K, et al. Glycan cross-feeding supports mutualism between Fusobacterium and the vaginal microbiota. PLoS Biol. 2020;18:e3000788.
    1. Gilbert NM, et al. Gardnerella vaginalis and Prevotella bivia trigger distinct and overlapping phenotypes in a mouse model of bacterial vaginosis. J. Infect. Dis. 2019;220:1099–1108.
    1. Uldbjerg N, Ekman G, Malmstrom A, Olsson K, Ulmsten U. Ripening of the human uterine cervix related to changes in collagen, glycosaminoglycans, and collagenolytic activity. Am. J. Obstet. Gynecol. 1983;147:662–666.
    1. Ketterer MR, et al. Desialylation of Neisseria gonorrhoeae lipooligosaccharide by cervicovaginal microbiome sialidases: the potential for enhancing infectivity in men. J. Infect. Dis. 2016;214:1621–1628.
    1. Vadillo-Ortega F, Estrada-Gutierrez G. Role of matrix metalloproteinases in preterm labour. BJOG. 2005;112(Suppl. 1):19–22.
    1. Athayde N, et al. Matrix metalloproteinases-9 in preterm and term human parturition. J. Matern. Fetal Med. 1999;8:213–219.
    1. Becher N, Hein M, Danielsen CC, Uldbjerg N. Matrix metalloproteinases in the cervical mucus plug in relation to gestational age, plug compartment, and preterm labor. Reprod. Biol. Endocrinol. 2010;8:113.
    1. Matheus ER, et al. MMP-9 expression increases according to the grade of squamous intraepithelial lesion in cervical smears. Diagn. Cytopathol. 2014;42:827–833.
    1. Zhang G, Miyake M, Lawton A, Goodison S, Rosser CJ. Matrix metalloproteinase-10 promotes tumor progression through regulation of angiogenic and apoptotic pathways in cervical tumors. BMC Cancer. 2014;14:310.
    1. Walther-Antonio MR, et al. Potential contribution of the uterine microbiome in the development of endometrial cancer. Genome Med. 2016;8:122.
    1. Łaniewski P, Ilhan ZE, Herbst-Kralovetz MM. The microbiome and gynaecological cancer development, prevention and therapy. Nat. Rev. Urol. 2020;17:232–250.
    1. Ravel J, et al. Daily temporal dynamics of vaginal microbiota before, during and after episodes of bacterial vaginosis. Microbiome. 2013;1:29.
    1. Anahtar MN, et al. Cervicovaginal bacteria are a major modulator of host inflammatory responses in the female genital tract. Immunity. 2015;42:965–976.
    1. Garcia, E. M., Kraskauskiene, V., Koblinski, J. E. & Jefferson, K. K. Interaction of Gardnerella vaginalis and vaginolysin with the apical versus basolateral face of a three-dimensional model of vaginal epithelium. Infect. Immun. 10.1128/IAI.00646-18 (2019).
    1. Eade CR, et al. Identification and characterization of bacterial vaginosis-associated pathogens using a comprehensive cervical-vaginal epithelial coculture assay. PLoS ONE. 2012;7:e50106.
    1. Łaniewski, P., Gomez, A., Hire, G., So, M. & Herbst-Kralovetz, M. M. Human three-dimensional endometrial epithelial cell model to study host interactions with vaginal bacteria and Neisseria gonorrhoeae. Infect. Immun. 10.1128/IAI.01049-16 (2017).
    1. Chan JF, et al. First report of spontaneous intrapartum Atopobium vaginae bacteremia. J. Clin. Microbiol. 2012;50:2525–2528.
    1. Gondwe T, et al. Novel bacterial vaginosis-associated organisms mediate the relationship between vaginal douching and pelvic inflammatory disease. Sex. Transm. Infect. 2020;96:439–444.
    1. Manhart LE, et al. Bacterial vaginosis-associated bacteria in men: association of Leptotrichia/Sneathia spp. with nongonococcal urethritis. Sex. Transm. Dis. 2013;40:944–949.
    1. Shukla SK, Meier PR, Mitchell PD, Frank DN, Reed KD. Leptotrichia amnionii sp. nov., a novel bacterium isolated from the amniotic fluid of a woman after intrauterine fetal demise. J. Clin. Microbiol. 2002;40:3346–3349.
    1. Decroix V, Goudjil S, Kongolo G, Mammeri H. ‘Leptotrichia amnionii’, a newly reported cause of early onset neonatal meningitis. J. Med. Microbiol. 2013;62:785–788.
    1. Passmore JA, Jaspan HB, Masson L. Genital inflammation, immune activation and risk of sexual HIV acquisition. Curr. Opin. HIV AIDS. 2016;11:156–162.
    1. Muenzner P, Hauck CR. Neisseria gonorrhoeae blocks epithelial exfoliation by nitric-oxide-mediated metabolic cross talk to promote colonization in mice. Cell Host Microbe. 2020;27:793–808 e795.
    1. Edwards JL. Neisseria gonorrhoeae survival during primary human cervical epithelial cell infection requires nitric oxide and is augmented by progesterone. Infect. Immun. 2010;78:1202–1213.
    1. Facchinetti F, Venturini P, Blasi I, Giannella L. Changes in the cervical competence in preterm labour. BJOG. 2005;112(Suppl. 1):23–27.
    1. Han J, et al. Elevated d-2-hydroxyglutarate during colitis drives progression to colorectal cancer. Proc. Natl Acad. Sci. USA. 2018;115:1057–1062.
    1. Ilhan ZE, et al. Deciphering the complex interplay between microbiota, HPV, inflammation and cancer through cervicovaginal metabolic profiling. EBioMedicine. 2019;44:675–690.
    1. Plummer EL, et al. Gardnerella vaginalis clade distribution is associated with behavioral practices and nugent score in women who have sex with women. J. Infect. Dis. 2020;221:454–463.
    1. Castro J, et al. Comparative transcriptomic analysis of Gardnerella vaginalis biofilms vs. planktonic cultures using RNA-seq. NPJ Biofilms Microbiomes. 2017;3:3.
    1. Herbst-Kralovetz MM, et al. Quantification and comparison of toll-like receptor expression and responsiveness in primary and immortalized human female lower genital tract epithelia. Am. J. Reprod. Immunol. 2008;59:212–224.
    1. Metsalu T, Vilo J. ClustVis: a web tool for visualizing clustering of multivariate data using principal component analysis and heatmap. Nucleic Acids Res. 2015;43:W566–W570.

Source: PubMed

3
Abonner