Next Article in Journal
Discovering Deleterious Single Nucleotide Polymorphisms of Human AKT1 Oncogene: An In Silico Study
Previous Article in Journal
Struggling with COVID-19 in Adult Inborn Errors of Immunity Patients: A Case Series of Combination Therapy and Multiple Lines of Therapy for Selected Patients
Previous Article in Special Issue
An Overview of the Microbiota of the Human Urinary Tract in Health and Disease: Current Issues and Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Crosstalk between the Resident Microbiota and the Immune Cells Regulates Female Genital Tract Health

by
Luigi Santacroce
1,*,†,
Raffaele Palmirotta
1,†,
Lucrezia Bottalico
2,
Ioannis Alexandros Charitos
3,
Marica Colella
1,
Skender Topi
2 and
Emilio Jirillo
1
1
Microbiology and Virology Section, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
2
Department of Clinical Disciplines, School of Technical Medical Sciences, “Alexander Xhuvani” University of Elbasan, 3001 Elbasan, Albania
3
Respiratory Rehabilitation Unit, Clinical Scientific Institutes Maugeri (IRCCS), 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Life 2023, 13(7), 1531; https://doi.org/10.3390/life13071531
Submission received: 7 May 2023 / Revised: 28 June 2023 / Accepted: 6 July 2023 / Published: 9 July 2023
(This article belongs to the Special Issue Urinary Microbiome and Genitourinary System Disorders)

Abstract

:
The female genital tract (FGT) performs several functions related to reproduction, but due to its direct exposure to the external environment, it may suffer microbial infections. Both the upper (uterus and cervix) and lower (vagina) FGT are covered by an epithelium, and contain immune cells (macrophages, dendritic cells, T and B lymphocytes) that afford a robust protection to the host. Its upper and the lower part differ in terms of Lactobacillus spp., which are dominant in the vagina. An alteration of the physiological equilibrium between the local microbiota and immune cells leads to a condition of dysbiosis which, in turn, may account for the outcome of FGT infection. Aerobic vaginitis, bacterial vaginosis, and Chlamydia trachomatis are the most frequent infections, and can lead to severe complications in reproduction and pregnancy. The use of natural products, such as probiotics, polyphenols, and lactoferrin in the course of FGT infections is an issue of current investigation. In spite of positive results, more research is needed to define the most appropriate administration, according to the type of patient.

1. Introduction

The female genital tract (FGT) consists of the uterine cervix, fallopian tubes, vagina and vulva, which perform various functional steps, such as the passage of the spermatozoa during insemination, and the expulsion of the fetus and placenta during delivery, and of the exfoliated endometrium during the menstrual period [1,2].
Because of its direct exposure to the external environment, the FGT is very susceptible to suffering microbial infections and inflammation. Under steady-state conditions, protection from invading pathogens is afforded by epithelial cells, resident immune cells, and the microbiota [3]. The cervix and vagina are covered by a stratified squamous epithelium, which represents a barrier against pathogens, as demonstrated by the evidence that its destruction increases the risk of HIV infection [4]. Innate immunity is based on the function of uterine natural killer (NK) cells, which recognize and lyse infected cells [5].
Macrophages represent the second most copious contingent of immune cells in the endometrium, which engulf and kill microbes, as well as self-cells [6].
As far as adaptive immunity is concerned, it is mostly T cells that are found in cervical and vaginal tissues, with tissue-resident memory (TREM) cells protecting against pathogens [7].
The cervico-vaginal microbiota constitute a complex micro-ecosystem, which play a critical role in maintaining homeostasis. The cervico-vaginal microbiota and their main functions have been well known for a long time, thanks to Albert Döderlein’s 1892 work that discovered and described the main features of certain vaginal acidogenic bacteria. However, the uterine microbiota have been less investigated than those of the vagina. Their composition appears to depend on disease status, such as in the case of endometrial cancer, and implantation failure [8,9].
Lactobacillus spp. form the predominant genus in the healthy FGT, providing several beneficial effects to the host. In fact, they maintain the vaginal pH at 3.8–4.5, through the production of lactic acid by the vaginal epithelial cells. In turn, the lactic acid regulates inflammation in the acidic microenvironment, while inhibiting the proliferation of other microorganisms, and competing with pathogens for survival and nutrition.
According to a few reports, Lactobacillus spp. Are not often detected in the endometrium, where a prevalence of Gardnerella (G.) vaginalis, Enterobacter, and Streptococcus (S.) agalactiae has been observed [10,11,12,13,14,15].
In the vagina, the microbiota are influenced by hygienic conditions, temporal dynamics, menstrual cycles, hormone levels, and concurrent diseases [16,17].
An alteration of the FGT microbiota may lead to a condition of so-called dysbiosis, with an increase in pathogenic bacteria over Lactobacillus [18,19,20]. In this respect, the main dysbiosis-mediated infections include vaginitis, endometritis, cervicitis, and pelvic inflammatory disease (PID) [21,22,23].
Aerobic vaginitis (AV), bacterial vaginosis (BV), and Chlamydia trachomatis (C. t.) infections represent the most-studied FGT infections.
In women of childbearing age, the incidence of AV is about 10%, with a prevalence of aerobic bacteria or Enterococcus [24]. BV is the most frequent vaginal disease in childbearing-age women, with serious consequences, such as infertility, miscarriage, the premature rupture of membranes, premature delivery, and an increased risk of sexually transmitted infections [25,26]. Microbiologically, BV is characterized by a decrease in Lactobacillus and an increase in anaerobic and microaerobic bacteria [27]. The prevalence of C. t. in women aged 15–49 years is 1.5–7%, with an asymptomatic clinical course. As lactic acid is an inhibitor of C. t. growth, vaginal microbiota containing Lactobacillus iners, which produces less lactic acid, promote the risk of C. t. infection [28,29]. Among 10% of C. t.-infected women, a progression toward PID has been reported, with the risk of severe ectopic pregnancy, abnormal reproductive function, and cancer [30].
Novel therapeutic approaches to improve the function of the FGT immunity–microbiota axis are mainly represented by the administration of natural compounds; i.e., probiotics, lactoferrin (LF), and polyphenols.
Probiotics exert several beneficial effects, even including the release of antimicrobial substances, and the modulation of the immune system [31,32,33]. LF is an antimicrobial peptide (AMP) that is able to prevent bacterial and fungal infections in the FGT [34]. Polyphenols are endowed with antioxidant, anti-inflammatory, and microbicidal activities [35], but data related to their effects on FGT infections are still experimental.
In the present review, an emphasis will be placed on the description of the resident immune cells and the microbiota of the FGT. Then, the main dysbiosis-mediated FGT infections will be elucidated. Finally, novel therapeutic attempts to treat FGT infections with natural products will be discussed.

2. Anatomy of the Female Genital Tract

The FGT consists of the lower FGT (vagina and ectocervix), with a large colony of microbes, and the upper FGT (endocervix, uterus, and oviduct) which, conversely, is sterile. Histologically, the cervix is composed of stroma and epithelial cells with an extracellular matrix, including type I and type II collagen. Type I collagen, which represents 70% of the extracellular matrix, maintains tissue integrity. The endocervix is covered by a columnar epithelium, while the ectocervix and vagina are covered by continuous stratified non-keratinized squamous epithelial cells. The area between the endocervix and the ectocervix is the so-called transformation zone, which is composed of squamous columnar epithelial cells. Columnar epithelial cells are composed of tight junctions that are regulated by estrogen, cytokines, and growth factors. On the other hand, the squamous epithelium contains adhesive junctions and desmosome junctions, which permit the passage of small molecules through the intercellular space. Notably, epithelial cells can act as presenting cells, thus recognizing microbial antigens and secreting antibiotics, cytokines, and chemokines.

3. Main Features of the Cervical Mucus

Cervical mucus is a product of goblet/secretory cells, in the context of the crypts. Mucins are the main constituents of cervical mucus, with mainly MUC5B and MUC5AC contained in the cervix. MUC5B reaches its highest level at the ovulation phase, with a watery secretion, which permits the entry of sperm into the upper reproductive tract. Conversely, during the luteal phase, the MUC5B expression level is decreased by progesterone, thus leading to a thickening in the cervical mucus. There is evidence that the menstrual cycle determines a bimodal distribution of the cervical immune components. Taking into consideration IgG/IgA, lactoferrin, interleukin (IL)-10, and antimicrobial peptides, these factors are expressed at higher levels at the early stage of the follicular phase, followed by a dramatic decrease in the middle of menstruation, and a further increase at the latter stage of menstruation.
In general terms, the cervical mucus represents a defense barrier, with mucin able to capture pathogens; meanwhile, the local immune response begins to act. In this last respect, lactoferrin, IgG, and IgA inhibit the adhesion and invasion of the cervical cells by pathogenic microorganisms [36].

4. The Immune Arsenal of the FGT

4.1. Innate Immunity

The epithelial cells of the FGT are non-immune cells that mostly defend the lower reproductive tract, cervix, and vagina. A stratified squamous epithelium, where tight junctions (TJ) predominate, covers the lower tract, while the upper portion, fallopian tubes, uterus, and inner cervix are characterized by a monolayer columnar epithelium, endowed with a more tightly connected network [37,38]. The effectiveness of the epithelial barrier of the FGT is supported by the evidence that its destruction appears to promote HIV infection, as well as the recruitment of HIV target cells. As was recently reported, the treatment of bovine endometrial epithelial cell lines with astaxanthin, a carotenoid, could reduce the lipopolysaccharide (LPS)-induced release of pro-inflammatory cytokines, while increasing the activity of cell superoxide dismutase and catalase, insulin-like growth factor, and epithelial growth factor [39]. In particular, such a treatment led to an increased expression of claudin, a TJ involved in the maintenance of the epithelial defense barrier against pathogen invasion.
Thanks to the toll-like receptors (TLRs) expressed on their membrane, epithelial cells from FGT are able to bind specific molecules of gram-positive and gram-negative bacteria, e.g., peptidoglycans and LPS, respectively [40], thus secreting anti-microbial peptides (AMPs) and cytokines [41,42,43,44]. AMPs, such as human beta defensin (HBD), LL-37, S200 protein, lysozyme, and iron metabolism proteins, prevent invasion by microbes in the upper FGT, while regulating the vaginal microbiota [45].
Moreover, epithelial cells from the FGT release a plethora of cytokines, i.e., interleukin (IL)-1 alpha, IL-1 beta, and tumor necrosis factor (TNF)-alpha, in response to Prevotella, Mobiluncus, and Sneathia, in the course of BV [46,47].
Natural killer (NK) cells are mostly confined to the uterus and perform a protective function against pathogens [48]. Functionally, NK cells protect the placenta from Listeria infection, and prevent the infection-induced abortion via secretion of granulysin into the placental trophoblast [49].
Macrophages are very abundant in the endometrium, playing a double role in the context of the FGT [50]. In fact, they engulf and kill pathogens, thus contributing to local innate immune defenses [51]. On the other hand, they maintain a tolerance milieu in the FGT, via the release of interleukin (IL)-10, transforming growth factor (TGF)-beta, and indoleamine (IDO) [52,53].
Dendritic cells (DCs) are the major APCs of the immune system and, via the activation of T regulatory (TREG) cells, maintain a steady-state condition in the FGT microenvironment [54]. Conversely, evidence has been provided that in the context of the cervical mucosa, DCs can capture and transfer HIV-1 via Siglec-1, thus facilitating viral dissemination [55].

4.2. Adaptive Immunity

In the context of the FGT, T and B lymphocytes represent the major players. In the human cervical mucosa and vagina, antigen-specific TRM CD8+ cells have been identified, and T-cell-induced vaccines are able to prevent FGT HIV infection [56,57]. Quite interestingly, estradiol has been shown to increase the TRM CD4+ cell response to the Herpes simplex (HSV) virus, via IL-17 secretion [58]. With special reference to the contingent of FGT Treg cells, they maintain the local immune tolerance, also attenuating the inflammatory response in the case of microbial stimulation of the immune system [59].
B cells are scarcely present in the FGT mucosa and, just recently, it has been reported that migrant memory B cells secrete specific IgG-2b anti-HIV in the vagina [60].
The immune components of the FGT are described in Figure 1.
Studies on the effects of the menstrual cycle and menopause on cervical T lymphocytes are controversial. For instance, the total number of CD8 + T cells and CD4 + T cells in the cervix remains unaltered, even including CD8 + T cell toxicity. Notably, transforming growth factor (TGF)-beta plays a dual role, inhibiting endometrial CD8 + T cells in the secretory phase, while TGF-beta does not act on cervical CD8 + T cells. This still allows a protective role by the cervix against pathogens, when the secretory endometrium is immunosuppressed by embryo implantation. Conversely, other studies report that the menstrual cycle and age impair the immune function. The levels of chemokine (C-C) motif ligand 2 (CCL2) and local CD4+ TRM cells increase during the follicular phase, because the inhibitory effect of progesterone decreases, and more CD8+ TRM cells are recruited to the cervix.
Furthermore, the frequency of CD8 + T cells and DCs decreases with age in the cervical tissue of postmenopausal women, thus supporting the increased susceptibility of these women to genital infections.

5. Composition and Function of the FGT Microbiota

With special reference to microbial communities, there are differences between the upper and lower tract of the FGT, with a higher bacterial load in the latter [61,62]. Mostly, the vaginal microbiota have been the object of intensive investigation, which has led to the identification of five separate community state types (CSTs) [63,64,65]. Lactobacillus represents the major microbiota of the vagina, with Lactobacillus (L.) crispatus, L. gasseri, L. iners, and L. jensenii very dominant [66]. Notably, the microbiota that are non-Lactobacillus prevalent appear to be associated with disease status and inflammation [67].
Among CSTs, CST-I, CST-II, and CST-V, which contain higher levels of L. crispatus, L. gasseri, L. iners, and L. jensenii, are characterized by a low pH, a high concentration of lactic acid, and a less inflammatory status [68,69,70]. Conversely, CST-IV, dominated by bacteria such as Atopobium, G. vaginalis, Mobiluncus, Prevotella, Anaerococcus, and Sneathia, are somewhat associated with BV [71,72].
The composition of the vaginal microbiota seems to influence the epithelial barrier function, as in the case of L. crispatus, which is associated with mucus that is able to trap HIV more effectively than other types of Lactobacillus spp. do [67]. On the other hand, BV-associated bacteria produce sialidase, which degrades sialic acid, a critical component in the mucus [68].
The vaginal levels of AMPs are associated with vaginal microbial diversity, and higher amounts of HBD-2, lactoferrin (LF), and LL-37 have been detected in the cervico-vaginal lavage of women with BV [73,74,75].
It is worth mentioning the role played by microbial metabolites in the vagina. The levels of D-lactic acid (D-LA) and L-lactic acid (L-LA) produced by the vaginal Lactobacillus greatly contribute to vaginal health. In fact, both reduce the pH of the vaginal mucosa, preventing the growth of pathogens, and also reducing the release of pro-inflammatory cytokines by epithelial cells [76]. In particular, LA in vitro induced the release of the anti-inflammatory cytokine, Il-1 receptor antagonist, and inhibited the TLR-mediated production of pro-inflammatory cytokines, in response to seminal plasma [77]. Furthermore, Lactobacillus spp. produce hydrogen peroxide and bacteriocins, which inhibit the growth of pathogens [78,79]. Furthermore, cervicovaginal supernatants of L crispatus could restore the endocervical barrier integrity, otherwise impaired by G. vaginalis culture supernatants [80].
A few studies have been focused on the role of vaginal short-chain fatty acids (SCFAs), for their protective role at the gut-mucosa level [81]. There is evidence that SCFAs are elevated in the vaginal mucosa of BV patients [82].
According to another report, high levels of SCFAs could, in vitro, trigger the release of pro-inflammatory cytokines from vaginal and cervical epithelial cells [83]. Taken together, the above results suggest that more studies are required, to clarify the exact role of SCFAs in the FGT.
The composition and function of the FGT microbiota are illustrated in Figure 2.

6. Dysbiosis and FGT Infections

Both the microbiota and metabolites undergo modifications during cervicovaginal dysbiosis. Lactobacilli utilize sugars, such as glycogen and glycogen hydrolysates, to produce lactic acid that, together with bacteriocins and biosurfactants, protect the cervicovaginal milieu. As described in the following paragraphs, BV is one of the most frequent forms of dysbiosis in the vagina, due to a decrease in Lactobacilli and an increase in anaerobic bacteria. At the same time, metabolites are also altered during cervicovaginal dysbiosis, with high levels of biogenic amines and short-chain fatty acids, and low levels of some amino acids, such as tyrosine and glutamate. Conversely, the metabolic signature of AV is less defined than that of BV [84,85,86].
In the following paragraphs, the major FGT infections will be described, in relation to the condition of dysbiosis.

Aerobic Vaginitis

AV patients harbor increased numbers of aerobic bacteria or Enterococcus in the vagina. Among these, Escherichia (E.) coli, S. agalactiae, S. anginosus, Staphylococcus (S.) aureus, S. epidermidis, and Enterococcus faecalis are the main ones.
With special reference to the pathogenic mechanisms elicited by AV-related bacteria, Staphylococcus spp. in the vagina cause an increase in SCFAs, especially acetate, as well as the triggering of the anti-inflammatory pathway sustained by Treg cells [87,88,89]. Conversely, in AV, other uropathogens are able to increase the expression of an array of pro-inflammatory cytokines, even including IL-1 beta, IL-6, IL-8, and TNF-alpha, as an indication of the local immune imbalance between inflammatory and anti-inflammatory forces [90,91]. Quite interestingly, some bacteria, in the course of AV, become resistant to the host immune response, as in the case of S. aureus, which evade the microbicidal activity locally produced by nitric oxide (NO) [92,93]. Moreover, S. aureus upregulates the NO-inducible lactate dehydrogenase, thus surviving to lactic acid fermentation [94]. In the same manner, the toxic shock syndrome toxin-1 (TSST-1) strains elude the local immune response, and induce the release of pro-inflammatory cytokines from vaginal epithelial cells, with a severe damage of the mucosal barrier [95].
There is evidence that, in BV patients, the vaginal microbiota are subverted with a decrease in Lactobacillus spp. and an increase in anaerobic bacteria and microprobes, e.g., G. vaginalis, Atopobium, Mycoplasma, Megasphaera, Mobiluncus, Roseburia, Diadister, Sneathia, and Prevotella spp. [96,97]. Notably, in BV patients, the metabolites of the genital tract appear to be more specific indicators of the disease phenotype than the bacteria do [98]. For instance, 2-hydroxyisovalerate and gamma-hydroxybutyrate are increased in the course of BV, with a decrease in lactic acid and tyrosine [99]. Furthermore, the production of succinic acid by Prevotella spp. and Mobiluncus spp. leads to the inhibition of leukocyte chemotaxis, and the modulation of the immune response. Moreover, the release of SCFAs in combination with bacteria influences the vaginal immune response, either recruiting neutrophils and monocytes, or inhibiting the release of pro-inflammatory cytokines [100].
C. t. infection is widely diffused among women aged 15–49 years, and may progress toward severe complications, such as ectopic pregnancy, reproductive abnormalities, and cancer [101]. From a pathogenic point of view, the predominance of L. iners in the vagina increases the risk of C. t. infection, as it produces less lactic acid [102]. In this respect, the production of L. iners for therapeutic use has started [103].
Furthermore, in C. t.-infected patients, the vaginal levels of valine, isoleucine, tyramine, cadaverine, and succinate were lower than those detected in healthy people [104]. This may indicate that C. t. likely utilizes nitrogen as a nutrient source or affects the nitrogen metabolism of the host. INDO is largely produced by Prevotella spp. in the course of BV, and it may promote C. t. overgrowth [105,106].
C. t. exploits INDO, to activate the synthesis of tryptophan, thus neutralizing the inhibition of C. t. growth mediated by interferon (IFN)-gamma [102]. In fact, IFN-gamma promotes the synthesis of indoleamine 2,3-dioxygenase with the consumption of tryptophan required for the growth of C. t. [107].
Another mechanism of escape by C. t. relies on its property of depriving the inducible nitric oxide synthase (iNOS) substrate, arginin, ultimately abrogating the microbicidal activity of NO [108].
The evasion of the immune response by C. t. leads to a condition of chronic infection, where T helper (h)1, Th2, and Th17 lymphocytes become activated, provoking damage, fibrosis, and scarring of the FGT [109,110].
The pathogenic mechanisms of FGT infections are expressed in Table 1.

7. Treatment with Natural Products for Maintaining the Health of the FGT

As discussed in the previous sections of this review, the dysbiosis of the FGT leads to different pathologies, even including infectious events. In particular, the misuse and abuse of antibiotics reduces the Lactobacillus contingent in the vagina, promoting a resistance to antimicrobials, with the generation of multi-resistant microorganisms [111,112,113,114]. Therefore, attempts have been made to normalize the vaginal microbiota with the supplementation of natural products.
Among these products, probiotics are currently used in FGT infections [115]. By definition, probiotics are “live microorganisms that, when administered in adequate amounts, confer a health benefit to the host” [116].
As was recently reported [117,118], for the treatment of FGT infections, probiotics can be administered in various combinations, i.e., vaginal/oral capsules, vaginal/oral powders, ovules, and sanitary towels. As far as the mechanisms of action of probiotics are concerned, they include the production of antimicrobials (bacteriocins, lantibiotics) and enzymes (arginine deaminase), promotion of adherence to epithelial cells, release of the components of mucus, extracellular matrix-colonization permanence, competition for nutrients, and modulation of the immune system [119,120].
Among relevant clinical trials, evidence has been provided that the administration of four Lactobacillus species could increase the vaginal colonization of Lactobacillus, followed by an improvement in the clinical symptoms of the infection [121,122,123].
Concerning the local administration of probiotics, vaginal suppositories increased the total number of vaginal Lactobacillus bacteria after seven days of treatment, in view of their ability to adhere to, and colonize, the vaginal epithelium [124]. A Lactin-V (L. CTV-05) vaginal treatment gave rise to a lower incidence of BV recurrence [125]. In a series of trials based on the administration of probiotic strains to healthy women, in patients with AV and BV, or in women with vulvo-vaginal candidiasis, an absence of side effects, the colonization and permanence of Lactobacillus, and reduced number of recurrences were observed [126,127,128].
Commercial products have been shown to be very effective in the course of FGT infections. Trivagin°, a product enriched in L. rhamnosus, L. gasseri, L. fermentum, and L. plantarum was very effective in normalizing the vaginal microbiota and attenuating symptoms of inflammation [129]. In another trial, women with BV who were treated with Ecovag° vaginal gelatin capsules, containing two strains of Lactobacillus, demonstrated resolution of disease at the end of the six-month follow-up [130]. Floridia® vaginal tablets, composed of L. brevis CD2, L. salivarius FV2, and L. plantarum FV9, were very effective in the eradication of BV-related bacteria, with a decrease in pro-inflammatory cytokines and reactive oxygen species (ROS) [131,132]. Notably, probiotic suppositories are usually devoid of major side effects, but vaginal discharge has been reported in some studies [113,119,120,133].
Despite the above-cited positive results, further trials are needed, to establish the efficacy of probiotics in pregnant and post-menopausal women, and for the prevention of preterm birth.
Polyphenols are natural substances that are largely contained in fruits, vegetables, cereals, extra virgin olive oil, and red wine [35,134]. They are endowed with antioxidant, anti-inflammatory, and antimicrobial effects and, therefore, are currently used in different pathologies [135,136]. With special reference to FGT infections, a recent paper has demonstrated the anti-inflammatory activity of polydatin, a polyphenol extracted from the rhizome of Polygonum cuspidatum, in the course of LPS-induced murine endometritis [137]. Polydatin was able to deactivate the NF-kB pathway, with a dramatic reduction in pro-inflammatory cytokine release. Therefore, it was proposed as a potential remedy in the case of human endometritis. A resveratrol-loaded liposome has been prepared for the topical treatment of vaginal inflammation and infections, in view of its in vitro scavenging activity, and its inhibition of pro-inflammatory cytokine release [138].
Lactoferrin is an AMP endowed with antimicrobial and immunomodulating properties, secreted by the uterine and vaginal epithelial cells [139,140,141]. The LF levels in cervicovaginal fluid increase in the course of FGT infections, correlating with the number of infiltrating neutrophils [142,143]. In a series of clinical trials in women with dysbiosis of the FGT and bacterial and fungal infections, the oral and/or intravaginal administration of LF led to clinical and histological improvements, with a dramatic reduction in pathogens and biomarkers of inflammation [144,145,146,147]. Of note, LF from bovine milk has been used as a dietary supplement, acting as a prebiotic on the intestinal microbiota, and increasing the colonization of Lactobacillus strains in the vagina [148].
The effects of natural-product administration on FGT infections are described in Table 2.

8. Conclusions

In summary, the network between the FGT microbiota and the local immune system plays a fundamental role in the maintenance of homeostasis, with special reference to the vaginal milieu. Any disturbance of the above indicated equilibrium may lead to a disease status; i.e., AV, BV, and C. t. infection, which may culminate in female infertility. The correction of FGT dysbiosis using natural products is a field of current interest, and clinical trials using probiotics and LF have led to positive results. Polyphenols, despite their well-known antioxidant, anti-inflammatory, and antimicrobial activities, have been investigated less.
In general, further data are needed, to define the exact interactions between FGT microbes and natural products, as well as in relation to the disease status of patients.

Funding

This paper has been supported by a grant from AIMS (Associazione Italiana Medici Specializzandi). The funders had no role in, or influence on, the content of the manuscript.

Authors Contributions

Concept and design: L.S. and E.J. Acquisition, analysis, and interpretation of data: R.P., M.C., and S.T. Drafting of the manuscript: I.A.C. and L.S. Critical revision of the manuscript for important intellectual content: all the authors. Administrative, technical, or material support: L.B. and L.S. Supervision: L.S. Project administration: L.S. and E.J. All authors have read and agreed to the published version of the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data for this manuscript have been reported in the paper.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMPsAntimicrobial peptides
APCsAntigen-presenting cells
AVAerobic vaginitis
BVBacterial vaginosis
CSTsCommunity state types
DCsDendritic cells
D-LAD-lactic acid
FGTFemale genital tract
HBDHuman beta defensin
HSVHerpes simplex virus
ILInterleukin
INOSInducible nitric oxide
LFLactoferrin
LPSLipopolysaccharide
L-LAL-lactic acid
NKNatural killer
PIDPelvic inflammatory disease
ROSReactive oxygen species
SCFAsShort-chain fatty acids
ThT helper
TGFTransforming growth factor
TJTight junction
TregT regulatory cell
TREMTissue resident memory
TSST-1Toxic shock syndrome toxin-1

References

  1. Rodriguez-Garcia, M.; Patel, M.V.; Shen, Z.; Bodwell, J.; Rossoll, R.M.; Wira, C.R. Tenofovir Inhibits Wound Healing of Epithelial Cells and Fibroblasts from the Upper and Lower Human Female Reproductive Tract. Sci. Rep. 2017, 8, 45725. [Google Scholar] [CrossRef] [Green Version]
  2. Gao, X.S.; Laven, J.; Louwers, Y.; Budding, A.; Schoenmakers, S. Microbiome as a predictor of implantation. Curr. Opin. Obstet. Gynecol. 2022, 34, 122–132. [Google Scholar] [CrossRef]
  3. Wang, Y.; Wang, X.; Zhu, M.; Ge, L.; Liu, X.; Su, K.; Chen, Z.; Zhao, W. The Interplay between Cervicovaginal Microbial Dysbiosis and Cervicovaginal Immunity. Front. Immunol. 2022, 13, 857299. [Google Scholar] [CrossRef]
  4. Mhlekude, B.; Lenman, A.; Sidoyi, P.; Joseph, J.; Kruppa, J.; Businge, C.B.; Mdaka, M.L.; Konietschke, F.; Pich, A.; Gerold, G.; et al. The barrier functions of crude cervical mucus plugs against HIV-1 infection in the context of cell-free and cell-to-cell transmission. AIDS 2021, 35, 2105–2117. [Google Scholar] [CrossRef]
  5. Le, T.; Reeves, R.K.; McKinnon, L.R. The functional diversity of tissue-resident natural killer cells against infection. Immunology 2022, 167, 28–39. [Google Scholar] [CrossRef]
  6. Dogra, P.; Farber, D.L. Stealth Killing by Uterine NK Cells for Tolerance and Tissue Homeostasis. Cell 2020, 182, 1074–1076. [Google Scholar] [CrossRef] [PubMed]
  7. Woodward Davis, A.S.; Vick, S.C.; Pattacini, L.; Voillet, V.; Hughes, S.M.; Lentz, G.M.; Kirby, A.C.; Fialkow, M.F.; Gottardo, R.; Hladik, F.; et al. The human memory T cell compartment changes across tissues of the female reproductive tract. Mucosal Immunol. 2021, 14, 862–872. [Google Scholar] [CrossRef] [PubMed]
  8. Baxter, N.T.; Ruffin, M.T., 4th; Rogers, M.A.; Schloss, P.D. Microbiota-based model improves the sensitivity of fecal immunochemical test for detecting colonic lesions. Genome Med. 2016, 8, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Ichiyama, T.; Kuroda, K.; Nagai, Y.; Urushiyama, D.; Ohno, M.; Yamaguchi, T.; Nagayoshi, M.; Sakuraba, Y.; Yamasaki, F.; Hata, K.; et al. Analysis of vaginal and endometrial microbiota communities in infertile women with a history of repeated implantation failure. Reprod. Med. Biol. 2021, 20, 334–344. [Google Scholar] [CrossRef] [PubMed]
  10. Møller, B.R.; Kristiansen, F.V.; Thorsen, P.; Frost, L.; Mogensen, S.C. Sterility of the uterine cavity. Acta Obstet. Gynecol. Scand. 1995, 74, 216–219. [Google Scholar] [CrossRef] [PubMed]
  11. Winters, A.D.; Romero, R.; Gervasi, M.; Gomez-Lopez, N.; Tran, M.R.; Garcia-Flores, V.; Pacora, P.; Jung, E.; Hassan, S.S.; Hsu, C.D.; et al. Does the endometrial cavity have a molecular microbial signature? Sci. Rep. 2019, 9, 9905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Moreno, I.; Codoñer, F.M.; Vilella, F.; Valbuena, D.; Martinez-Blanch, J.F.; Jimenez-Almazán, J.; Alonso, R.; Alamá, P.; Remohí, J.; Pellicer, A.; et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am. J. Obstet. Gynecol. 2016, 215, 684–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Toson, B.; Simon, C.; Moreno, I. The Endometrial Microbiome and Its Impact on Human Conception. Int. J. Mol. Sci. 2022, 23, 485. [Google Scholar] [CrossRef] [PubMed]
  14. Moreno, I.; Garcia-Grau, I.; Perez-Villaroya, D.; Gonzalez-Monfort, M.; Bahçeci, M.; Barrionuevo, M.J.; Taguchi, S.; Puente, E.; Dimattina, M.; Lim, M.W.; et al. Endometrial microbiota composition is associated with reproductive outcome in infertile patients. Microbiome 2022, 10, 1. [Google Scholar] [CrossRef]
  15. Moreno, I.; Garcia-Grau, I.; Bau, D.; Perez-Villaroya, D.; Gonzalez-Monfort, M.; Vilella, F.; Romero, R.; Simón, C. The first glimpse of the endometrial microbiota in early pregnancy. Am. J. Obstet. Gynecol. 2020, 222, 296–305. [Google Scholar] [CrossRef]
  16. Greenbaum, S.; Greenbaum, G.; Moran-Gilad, J.; Weintraub, A.Y. Ecological dynamics of the vaginal microbiome in relation to health and disease. Am. J. Obstet. Gynecol. 2019, 220, 324–335. [Google Scholar] [CrossRef]
  17. Gajer, P.; Brotman, R.M.; Bai, G.; Sakamoto, J.; Schütte, U.M.; Zhong, X.; Koenig, S.S.; Fu, L.; Ma, Z.S.; Zhou, X.; et al. Temporal dynamics of the human vaginal microbiota. Sci. Transl. Med. 2012, 4, 132ra52. [Google Scholar] [CrossRef] [Green Version]
  18. Lev-Sagie, A.; Goldman-Wohl, D.; Cohen, Y.; Dori-Bachash, M.; Leshem, A.; Mor, U.; Strahilevitz, J.; Moses, A.E.; Shapiro, H.; Yagel, S.; et al. Vaginal microbiome transplantation in women with intractable bacterial vaginosis. Nat. Med. 2019, 25, 1500–1504. [Google Scholar] [CrossRef]
  19. Torcia, M.G. Interplay among Vaginal Microbiome, Immune Response and Sexually Transmitted Viral Infections. Int. J. Mol. Sci. 2019, 20, 266. [Google Scholar] [CrossRef] [Green Version]
  20. d’Enfert, C.; Kaune, A.K.; Alaban, L.R.; Chakraborty, S.; Cole, N.; Delavy, M.; Kosmala, D.; Marsaux, B.; Fróis-Martins, R.; Morelli, M.; et al. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: Current knowledge and new perspectives. FEMS Microbiol. Rev. 2021, 45, fuaa060. [Google Scholar] [CrossRef]
  21. Sherrard, J.; Wilson, J.; Donders, G.; Mendling, W.; Jensen, J.S. 2018 European (IUSTI/WHO) International Union against sexually transmitted infections (IUSTI) World Health Organisation (WHO) guideline on the management of vaginal discharge. Int. J. STD AIDS 2018, 29, 1258–1272. [Google Scholar] [CrossRef]
  22. Workowski, K.A.; Bolan, G.A.; Centers for Disease Control and Prevention. Sexually transmitted diseases treatment guidelines, 2015. MMWR Recomm. Rep. 2015, 64, 1–137. [Google Scholar] [PubMed]
  23. Song, S.D.; Acharya, K.D.; Zhu, J.E.; Deveney, C.M.; Walther-Antonio, M.R.S.; Tetel, M.J.; Chia, N. Daily Vaginal Microbiota Fluctuations Associated with Natural Hormonal Cycle, Contraceptives, Diet, and Exercise. mSphere 2020, 5, e00593-20. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, C.; Fan, A.; Li, H.; Yan, Y.; Qi, W.; Wang, Y.; Han, C.; Xue, F. Vaginal bacterial profiles of aerobic vaginitis: A case-control study. Diagn. Microbiol. Infect. Dis. 2020, 96, 114981. [Google Scholar] [CrossRef]
  25. Bhatia, M.; Mitsi, V.; Court, L.; Thampi, P.; El-Nasharty, M.; Hesham, S.; Randall, W.; Davies, R.; Impey, L. The outcomes of pregnancies with reduced fetal movements: A retrospective cohort study. Acta Obstet. Gynecol. Scand. 2019, 98, 1450–1454. [Google Scholar] [CrossRef]
  26. Khelaifia, S.; Lagier, J.C.; Nkamga, V.D.; Guilhot, E.; Drancourt, M.; Raoult, D. Aerobic culture of methanogenic archaea without an external source of hydrogen. Eur. J. Clin. Microbiol. Infect. Dis. 2016, 35, 985–991. [Google Scholar] [CrossRef] [PubMed]
  27. Sabo, M.C.; Balkus, J.E.; Richardson, B.A.; Srinivasan, S.; Kimani, J.; Anzala, O.; Schwebke, J.; Feidler, T.L.; Fredricks, D.N.; McClelland, R.S. Association between vaginal washing and vaginal bacterial concentrations. PLoS ONE 2019, 14, e0210825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. van Houdt, R.; Ma, B.; Bruisten, S.M.; Speksnijder, A.G.C.L.; Ravel, J.; de Vries, H.J.C. Lactobacillus iners-dominated vaginal microbiota is associated with increased susceptibility to Chlamydia trachomatis infection in Dutch women: A case-control study. Sex. Transm. Infect. 2018, 94, 117–123. [Google Scholar] [CrossRef]
  29. Gargiulo Isacco, C.; Balzanelli, M.G.; Garzone, S.; Lorusso, M.; Inchingolo, F.; Nguyen, K.C.D.; Santacroce, L.; Mosca, A.; Del Prete, R. Alterations of Vaginal Microbiota and Chlamydia trachomatis as Crucial Co-Causative Factors in Cervical Cancer Genesis Procured by HPV. Microorganisms 2023, 11, 662. [Google Scholar] [CrossRef]
  30. Idahl, A.; Le Cornet, C.; González Maldonado, S.; Waterboer, T.; Bender, N.; Tjønneland, A.; Hansen, L.; Boutron-Ruault, M.C.; Fournier, A.; Kvaskoff, M.; et al. Serologic markers of Chlamydia trachomatis and other sexually transmitted infections and subsequent ovarian cancer risk: Results from the EPIC cohort. Int. J. Cancer 2020, 147, 2042–2052. [Google Scholar] [CrossRef] [Green Version]
  31. McFarland, L.V.; Evans, C.T.; Goldstein, E.J.C. Strain-Specificity and Disease-Specificity of Probiotic Efficacy: A Systematic Review and Meta-Analysis. Front. Med. 2018, 5, 124. [Google Scholar] [CrossRef] [Green Version]
  32. Reid, G. Probiotics: Definition, scope and mechanisms of action. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 17–25. [Google Scholar] [CrossRef] [PubMed]
  33. Nader-Macías, M.E.F.; De Gregorio, P.R.; Silva, J.A. Probiotic lactobacilli in formulas and hygiene products for the health of the urogenital tract. Pharmacol. Res. Perspect. 2021, 9, e00787. [Google Scholar] [CrossRef] [PubMed]
  34. Artym, J.; Zimecki, M.; Kruzel, M.L. Lactoferrin for Prevention and Treatment of Anemia and Inflammation in Pregnant Women: A Comprehensive Review. Biomedicines 2021, 9, 898. [Google Scholar] [CrossRef]
  35. Magrone, T.; Magrone, M.; Russo, M.A.; Jirillo, E. Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants 2019, 9, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Lacroix, G.; Gouyer, V.; Gottrand, F.; Desseyn, J.L. The Cervicovaginal Mucus Barrier. Int. J. Mol. Sci. 2020, 21, 8266. [Google Scholar] [CrossRef] [PubMed]
  37. Lee, S.K.; Kim, C.J.; Kim, D.J.; Kang, J.H. Immune cells in the female reproductive tract. Immune Netw. 2015, 15, 16–26. [Google Scholar] [CrossRef] [Green Version]
  38. Charitos, I.A.; Topi, S.; Gagliano-Candela, R.; De Nitto, E.; Polimeno, L.; Montagnani, M.; Santacroce, L. The Toxic Effects of Endocrine Disrupting Chemicals (EDCs) on Gut Microbiota: Bisphenol A (BPA) A Review. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 716–727. [Google Scholar] [CrossRef]
  39. Wan, F.C.; Zhang, C.; Jin, Q.; Wei, C.; Zhao, H.B.; Zhang, X.L.; You, W.; Liu, X.M.; Liu, G.F.; Liu, Y.F.; et al. Protective effects of astaxanthin on lipopolysaccharide-induced inflammation in bovine endometrial epithelial cells†. Biol. Reprod. 2020, 102, 339–347. [Google Scholar] [CrossRef]
  40. Benjelloun, F.; Quillay, H.; Cannou, C.; Marlin, R.; Madec, Y.; Fernandez, H.; Chrétien, F.; Le Grand, R.; Barré-Sinoussi, F.; Nugeyre, M.T.; et al. Activation of Toll-Like Receptors Differentially Modulates Inflammation in the Human Reproductive Tract: Preliminary Findings. Front. Immunol. 2020, 11, 1655. [Google Scholar] [CrossRef]
  41. Magrone, T.; Russo, M.A.; Jirillo, E. Dietary Approaches to Attain Fish Health with Special Reference to their Immune System. Curr. Pharm. Des. 2018, 24, 4921–4931. [Google Scholar] [CrossRef] [PubMed]
  42. Magrone, T.; Russo, M.A.; Jirillo, E. Antimicrobial Peptides in Human Disease: Therapeutic Approaches. Second of Two Parts. Curr. Pharm. Des. 2018, 24, 1148–1156. [Google Scholar] [CrossRef] [PubMed]
  43. Yarbrough, V.L.; Winkle, S.; Herbst-Kralovetz, M.M. Antimicrobial peptides in the female reproductive tract: A critical component of the mucosal immune barrier with physiological and clinical implications. Hum. Reprod. Update 2015, 21, 353–377. [Google Scholar] [CrossRef] [Green Version]
  44. Anahtar, M.N.; Gootenberg, D.B.; Mitchell, C.M.; Kwon, D.S. Cervicovaginal Microbiota and Reproductive Health: The Virtue of Simplicity. Cell Host Microbe 2018, 23, 159–168. [Google Scholar] [CrossRef] [Green Version]
  45. Yan, X.; Zhang, X.; Wu, H.H.; Wu, S.J.; Tang, X.Y.; Liu, T.Z.; Li, S. Novel T-cell signature based on cell pair algorithm predicts survival and immunotherapy response for patients with bladder urothelial carcinoma. Front. Immunol. 2022, 13, 994594. [Google Scholar] [CrossRef] [PubMed]
  46. Dong, M.; Dong, Y.; Bai, J.; Li, H.; Ma, X.; Li, B.; Wang, C.; Li, H.; Qi, W.; Wang, Y.; et al. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front. Cell. Infect. Microbiol. 2023, 13, 1124591. [Google Scholar] [CrossRef] [PubMed]
  47. Santacroce, L.; Sardaro, N.; Topi, S.; Pettini, F.; Bottalico, L.; Cantore, S.; Cascella, G.; Del Prete, R.; Dipalma, G.; Inchingolo, F. The pivotal role of oral microbiota in health and disease. J. Biol. Regul. Homeost. Agents 2020, 34, 733–737. [Google Scholar] [CrossRef]
  48. Bulmer, J.N.; Williams, P.J.; Lash, G.E. Immune cells in the placental bed. Int. J. Dev. Biol. 2010, 54, 281–294. [Google Scholar] [CrossRef]
  49. Weiss, G.; Schaible, U.E. Macrophage defense mechanisms against intracellular bacteria. Immunol. Rev. 2015, 264, 182–203. [Google Scholar] [CrossRef] [Green Version]
  50. Hussain, T.; Murtaza, G.; Kalhoro, D.H.; Kalhoro, M.S.; Yin, Y.; Chughtai, M.I.; Tan, B.; Yaseen, A.; Rehman, Z.U. Understanding the Immune System in Fetal Protection and Maternal Infections during Pregnancy. J. Immunol. Res. 2022, 2022, 7567708. [Google Scholar] [CrossRef]
  51. Santacroce, L.; Man, A.; Charitos, I.A.; Haxhirexha, K.; Topi, S. Current knowledge about the connection between health status and gut microbiota from birth to elderly. A narrative review. Front. Biosci. (Landmark Ed.) 2021, 26, 135–148. [Google Scholar] [CrossRef] [PubMed]
  52. Racicot, K.; Kwon, J.Y.; Aldo, P.; Silasi, M.; Mor, G. Understanding the complexity of the immune system during pregnancy. Am. J. Reprod. Immunol. 2014, 72, 107–116. [Google Scholar] [CrossRef] [PubMed]
  53. Perez-Zsolt, D.; Cantero-Pérez, J.; Erkizia, I.; Benet, S.; Pino, M.; Serra-Peinado, C.; Hernández-Gallego, A.; Castellví, J.; Tapia, G.; Arnau-Saz, V.; et al. Dendritic Cells From the Cervical Mucosa Capture and Transfer HIV-1 via Siglec-1. Front. Immunol. 2019, 10, 825. [Google Scholar] [CrossRef] [PubMed]
  54. Peng, T.; Phasouk, K.; Bossard, E.; Klock, A.; Jin, L.; Laing, K.J.; Johnston, C.; Williams, N.A.; Czartoski, J.L.; Varon, D.; et al. Distinct populations of antigen-specific tissue-resident CD8+ T cells in human cervix mucosa. JCI Insight 2021, 6, e149950. [Google Scholar] [CrossRef]
  55. Arunachalam, P.S.; Charles, T.P.; Joag, V.; Bollimpelli, V.S.; Scott, M.K.; Wimmers, F.; Burton, S.L.; Labranche, C.C.; Petitdemange, C.; Gangadhara, S.; et al. T cell-inducing vaccine durably prevents mucosal SHIV infection even with lower neutralizing antibody titers. Nat. Med. 2020, 26, 932–940. [Google Scholar] [CrossRef] [PubMed]
  56. Bagri, P.; Ghasemi, R.; McGrath, J.J.C.; Thayaparan, D.; Yu, E.; Brooks, A.G.; Stämpfli, M.R.; Kaushic, C. Estradiol Enhances Antiviral CD4+ Tissue-Resident Memory T Cell Responses following Mucosal Herpes Simplex Virus 2 Vaccination through an IL-17-Mediated Pathway. J. Virol. 2020, 95, e01206-20. [Google Scholar] [CrossRef] [PubMed]
  57. Tsuda, S.; Nakashima, A.; Morita, K.; Shima, T.; Yoneda, S.; Kishi, H.; Saito, S. The role of decidual regulatory T cells in the induction and maintenance of fetal antigen-specific tolerance: Imbalance between regulatory and cytotoxic T cells in pregnancy complications. Hum. Immunol. 2021, 82, 346–352. [Google Scholar] [CrossRef]
  58. Oh, J.E.; Iijima, N.; Song, E.; Lu, P.; Klein, J.; Jiang, R.; Kleinstein, S.H.; Iwasaki, A. Migrant memory B cells secrete luminal antibody in the vagina. Nature 2019, 571, 122–126. [Google Scholar] [CrossRef]
  59. Baker, J.M.; Chase, D.M.; Herbst-Kralovetz, M.M. Uterine Microbiota: Residents, Tourists, or Invaders? Front. Immunol. 2018, 9, 208. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, C.; Song, X.; Wei, W.; Zhong, H.; Dai, J.; Lan, Z.; Li, F.; Yu, X.; Feng, Q.; Wang, Z.; et al. The microbiota continuum along the female reproductive tract and its relation to uterine-related diseases. Nat. Commun. 2017, 8, 875. [Google Scholar] [CrossRef] [Green Version]
  61. Ravel, J.; Gajer, P.; Abdo, Z.; Schneider, G.M.; Koenig, S.S.; McCulle, S.L.; Karlebach, S.; Gorle, R.; Russell, J.; Tacket, C.O.; et al. Vaginal microbiome of reproductive-age women. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. 1), 4680–4687. [Google Scholar] [CrossRef] [PubMed]
  62. Ma, Z.S.; Li, L. Quantifying the human vaginal community state types (CSTs) with the species specificity index. PeerJ 2017, 5, e3366. [Google Scholar] [CrossRef] [Green Version]
  63. France, M.; Alizadeh, M.; Brown, S.; Ma, B.; Ravel, J. Towards a deeper understanding of the vaginal microbiota. Nat. Microbiol. 2022, 7, 367–378. [Google Scholar] [CrossRef] [PubMed]
  64. Ma, B.; Forney, L.J.; Ravel, J. Vaginal microbiome: Rethinking health and disease. Annu. Rev. Microbiol. 2012, 66, 371–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. De Seta, F.; Campisciano, G.; Zanotta, N.; Ricci, G.; Comar, M. The Vaginal Community State Types Microbiome-Immune Network as Key Factor for Bacterial Vaginosis and Aerobic Vaginitis. Front. Microbiol. 2019, 10, 2451. [Google Scholar] [CrossRef] [PubMed]
  66. Bommana, S.; Richards, G.; Kama, M.; Kodimerla, R.; Jijakli, K.; Read, T.D.; Dean, D. Metagenomic Shotgun Sequencing of Endocervical, Vaginal, and Rectal Samples among Fijian Women with and without Chlamydia trachomatis Reveals Disparate Microbial Populations and Function across Anatomic Sites: A Pilot Study. Microbiol. Spectr. 2022, 10, e0010522. [Google Scholar] [CrossRef]
  67. Schellenberg, J.J.; Links, M.G.; Hill, J.E.; Dumonceaux, T.J.; Kimani, J.; Jaoko, W.; Wachihi, C.; Mungai, J.N.; Peters, G.A.; Tyler, S.; et al. Molecular definition of vaginal microbiota in East African commercial sex workers. Appl. Environ. Microbiol. 2011, 77, 4066–4074. [Google Scholar] [CrossRef] [Green Version]
  68. Hedges, S.R.; Barrientes, F.; Desmond, R.A.; Schwebke, J.R. Local and systemic cytokine levels in relation to changes in vaginal flora. J. Infect. Dis. 2006, 193, 556–562. [Google Scholar] [CrossRef] [Green Version]
  69. Santacroce, L.; Bottalico, L.; Topi, S.; Castellaneta, F.; Charitos, I.A. The “Scourge of the Renaissance”. A Short Review about Treponema pallidum infection. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 335–343. [Google Scholar] [CrossRef]
  70. Nunn, K.L.; Wang, Y.Y.; Harit, D.; Humphrys, M.S.; Ma, B.; Cone, R.; Ravel, J.; Lai, S.K. Enhanced Trapping of HIV-1 by Human Cervicovaginal Mucus Is Associated with Lactobacillus crispatus-Dominant Microbiota. mBio 2015, 6, e01084-15. [Google Scholar] [CrossRef] [Green Version]
  71. Vagios, S.; Mitchell, C.M. Mutual Preservation: A Review of Interactions Between Cervicovaginal Mucus and Microbiota. Front. Cell. Infect. Microbiol. 2021, 11, 676114. [Google Scholar] [CrossRef]
  72. Borgdorff, H.; Gautam, R.; Armstrong, S.D.; Xia, D.; Ndayisaba, G.F.; van Teijlingen, N.H.; Geijtenbeek, T.B.; Wastling, J.M.; van de Wijgert, J.H. Cervicovaginal microbiome dysbiosis is associated with proteome changes related to alterations of the cervicovaginal mucosal barrier. Mucosal Immunol. 2016, 9, 621–633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Fan, S.R.; Liu, X.P.; Liao, Q.P. Human defensins and cytokines in vaginal lavage fluid of women with bacterial vaginosis. Int. J. Gynaecol. Obstet. 2008, 103, 50–54. [Google Scholar] [CrossRef] [PubMed]
  74. Gregory, M.S.; Hackett, C.G.; Abernathy, E.F.; Lee, K.S.; Saff, R.R.; Hohlbaum, A.M.; Moody, K.S.; Hobson, M.W.; Jones, A.; Kolovou, P.; et al. Opposing Roles for Membrane Bound and Soluble Fas Ligand in Glaucoma-Associated Retinal Ganglion Cell Death. PLoS ONE 2011, 6, e17659. [Google Scholar] [CrossRef] [Green Version]
  75. Hearps, A.C.; Tyssen, D.; Srbinovski, D.; Bayigga, L.; Diaz, D.J.D.; Aldunate, M.; Cone, R.A.; Gugasyan, R.; Anderson, D.J.; Tachedjian, G. Vaginal lactic acid elicits an anti-inflammatory response from human cervicovaginal epithelial cells and inhibits production of pro-inflammatory mediators associated with HIV acquisition. Mucosal Immunol. 2017, 10, 1480–1490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Vallor, A.C.; Antonio, M.A.; Hawes, S.E.; Hillier, S.L. Factors associated with acquisition of, or persistent colonization by, vaginal lactobacilli: Role of hydrogen peroxide production. J. Infect. Dis. 2001, 184, 1431–1436. [Google Scholar] [CrossRef] [Green Version]
  77. Chopra, C.; Bhushan, I.; Mehta, M.; Koushal, T.; Gupta, A.; Sharma, S.; Kumar, M.; Khodor, S.A.; Sharma, S. Vaginal microbiome: Considerations for reproductive health. Future Microbiol. 2022, 17, 1501–1513. [Google Scholar] [CrossRef] [PubMed]
  78. Anton, L.; Sierra, L.J.; DeVine, A.; Barila, G.; Heiser, L.; Brown, A.G.; Elovitz, M.A. Common Cervicovaginal Microbial Supernatants Alter Cervical Epithelial Function: Mechanisms by Which Lactobacillus crispatus Contributes to Cervical Health. Front. Microbiol. 2018, 9, 2181. [Google Scholar] [CrossRef] [Green Version]
  79. Blacher, E.; Levy, M.; Tatirovsky, E.; Elinav, E. Microbiome-Modulated Metabolites at the Interface of Host Immunity. J. Immunol. (Baltim. Md. 1950) 2017, 198, 572–580. [Google Scholar] [CrossRef] [Green Version]
  80. Mirmonsef, P.; Gilbert, D.; Zariffard, M.R.; Hamaker, B.R.; Kaur, A.; Landay, A.L.; Spear, G.T. The effects of commensal bacteria on innate immune responses in the female genital tract. Am. J. Reprod. Immunol. 2011, 65, 190–195. [Google Scholar] [CrossRef] [Green Version]
  81. Moosa, Y.; Kwon, D.; de Oliveira, T.; Wong, E.B. Determinants of Vaginal Microbiota Composition. Front. Cell. Infect. Microbiol. 2020, 10, 467. [Google Scholar] [CrossRef]
  82. Medina-Bastidas, D.; Camacho-Arroyo, I.; García-Gómez, E. Current findings in endometrial microbiome: Impact on uterine diseases. Reproduction 2022, 163, R81–R96. [Google Scholar] [CrossRef] [PubMed]
  83. Tortelli, B.A.; Lewis, W.G.; Allsworth, J.E.; Member-Meneh, N.; Foster, L.R.; Reno, H.E.; Peipert, J.F.; Fay, J.C.; Lewis, A.L. Associations between the vaginal microbiome and Candida colonization in women of reproductive age. Am. J. Obstet. Gynecol. 2020, 222, 471.e1–471.e9. [Google Scholar] [CrossRef] [PubMed]
  84. Hocking, J.S.; Geisler, W.M.; Kong, F.Y.S. Update on the Epidemiology, Screening, and Management of Chlamydia trachomatis Infection. Infect. Dis. Clin. N. Am. 2023, 37, 267–288. [Google Scholar] [CrossRef]
  85. Xiao, B.; Qin, H.; Mi, L.; Zhang, D. Correlation Analysis of Vaginal Microbiome Changes and Bacterial Vaginosis Plus Vulvovaginal Candidiasis Mixed Vaginitis Prognosis. Front. Cell. Infect. Microbiol. 2022, 12, 860589. [Google Scholar] [CrossRef] [PubMed]
  86. Olive, A.J.; Sassetti, C.M. Metabolic crosstalk between host and pathogen: Sensing, adapting and competing. Nat. Rev. Microbiol. 2016, 14, 221–234. [Google Scholar] [CrossRef]
  87. Postler, T.S.; Ghosh, S. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab. 2017, 26, 110–130. [Google Scholar] [CrossRef] [Green Version]
  88. Benner, M.; Ferwerda, G.; Joosten, I.; van der Molen, R.G. How uterine microbiota might be responsible for a receptive, fertile endometrium. Hum. Reprod. Update 2018, 24, 393–415. [Google Scholar] [CrossRef] [Green Version]
  89. Budilovskaya, O.V.; Shipitsina, E.V.; Spasibova, E.V.; Pereverzeva, N.A.; Vorob’eva, N.E.; Tsypurdeeva, N.D.; Grigoryev, A.N.; Savicheva, A.M. Differential Expression of Local Immune Response Genes in the Vagina: Implication for the Diagnosis of Vaginal Infections. Bull. Exp. Biol. Med. 2020, 168, 646–650. [Google Scholar] [CrossRef]
  90. Wagner, B.K.; Dey, M. Chemical strategies to understand and manipulate host-pathogen interactions. Cell Chem. Biol. 2022, 29, 711–712. [Google Scholar] [CrossRef]
  91. Santacroce, L.; Colella, M.; Charitos, I.A.; Di Domenico, M.; Palmirotta, R.; Jirillo, E. Microbial and Host Metabolites at the Backstage of Fever: Current Knowledge about the Co-Ordinate Action of Receptors and Molecules Underlying Pathophysiology and Clinical Implications. Metabolites 2023, 13, 461. [Google Scholar] [CrossRef]
  92. Richardson, A.R.; Libby, S.J.; Fang, F.C. A nitric oxide-inducible lactate dehydrogenase enables Staphylococcus aureus to resist innate immunity. Science 2008, 319, 1672–1676. [Google Scholar] [CrossRef] [PubMed]
  93. Pereira, N.; Edlind, T.D.; Schlievert, P.M.; Nyirjesy, P. Vaginal toxic shock reaction triggering desquamative inflammatory vaginitis. J. Low. Genit. Tract Dis. 2013, 17, 88–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Srinivasan, S.; Morgan, M.T.; Fiedler, T.L.; Djukovic, D.; Hoffman, N.G.; Raftery, D.; Marrazzo, J.M.; Fredricks, D.N. Metabolic signatures of bacterial vaginosis. mBio 2015, 6, e00204-15. [Google Scholar] [CrossRef] [Green Version]
  95. Chen, X.; Lu, Y.; Chen, T.; Li, R. The Female Vaginal Microbiome in Health and Bacterial Vaginosis. Front. Cell. Infect. Microbiol. 2021, 11, 631972. [Google Scholar] [CrossRef]
  96. Ceccarani, C.; Foschi, C.; Parolin, C.; D’Antuono, A.; Gaspari, V.; Consolandi, C.; Laghi, L.; Camboni, T.; Vitali, B.; Severgnini, M.; et al. Diversity of vaginal microbiome and metabolome during genital infections. Sci. Rep. 2019, 9, 14095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Yeoman, C.J.; Thomas, S.M.; Miller, M.E.; Ulanov, A.V.; Torralba, M.; Lucas, S.; Gillis, M.; Cregger, M.; Gomez, A.; Ho, M.; et al. A multi-omic systems-based approach reveals metabolic markers of bacterial vaginosis and insight into the disease. PLoS ONE 2013, 8, e56111. [Google Scholar] [CrossRef]
  98. McMillan, A.; Rulisa, S.; Sumarah, M.; Macklaim, J.M.; Renaud, J.; Bisanz, J.E.; Gloor, G.B.; Reid, G. A multi-platform metabolomics approach identifies highly specific biomarkers of bacterial diversity in the vagina of pregnant and non-pregnant women. Sci. Rep. 2015, 5, 14174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Al-Mushrif, S.; Eley, A.; Jones, B.M. Inhibition of chemotaxis by organic acids from anaerobes may prevent a purulent response in bacterial vaginosis. J. Med. Microbiol. 2000, 49, 1023–1030. [Google Scholar] [CrossRef] [Green Version]
  100. Vitali, B.; Cruciani, F.; Picone, G.; Parolin, C.; Donders, G.; Laghi, L. Vaginal microbiome and metabolome highlight specific signatures of bacterial vaginosis. Eur. J. Clin. Microbiol. Infect. Dis. 2015, 34, 2367–2376. [Google Scholar] [CrossRef]
  101. Edwards, V.L.; Smith, S.B.; McComb, E.J.; Tamarelle, J.; Ma, B.; Humphrys, M.S.; Gajer, P.; Gwilliam, K.; Schaefer, A.M.; Lai, S.K.; et al. The Cervicovaginal Microbiota-Host Interaction Modulates Chlamydia trachomatis Infection. mBio. 2019, 10, e01548-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Parolin, C.; Foschi, C.; Laghi, L.; Zhu, C.; Banzola, N.; Gaspari, V.; D’Antuono, A.; Giordani, B.; Severgnini, M.; Consolandi, C.; et al. Insights Into Vaginal Bacterial Communities and Metabolic Profiles of Chlamydia trachomatis Infection: Positioning Between Eubiosis and Dysbiosis. Front. Microbiol. 2018, 9, 600. [Google Scholar] [CrossRef] [PubMed]
  103. Borgdorff, H.; Armstrong, S.D.; Tytgat, H.L.; Xia, D.; Ndayisaba, G.F.; Wastling, J.M.; van de Wijgert, J.H. Unique Insights in the Cervicovaginal Lactobacillus iners and L. crispatus Proteomes and Their Associations with Microbiota Dysbiosis. PLoS ONE 2016, 11, e0150767. [Google Scholar] [CrossRef]
  104. Sasaki-Imamura, T.; Yoshida, Y.; Suwabe, K.; Yoshimura, F.; Kato, H. Molecular basis of indole production catalyzed by tryptophanase in the genus Prevotella. FEMS Microbiol. Lett. 2011, 322, 51–59. [Google Scholar] [CrossRef] [Green Version]
  105. Lewis, M.E.; Belland, R.J.; AbdelRahman, Y.M.; Beatty, W.L.; Aiyar, A.A.; Zea, A.H.; Greene, S.J.; Marrero, L.; Buckner, L.R.; Tate, D.J.; et al. Morphologic and molecular evaluation of Chlamydia trachomatis growth in human endocervix reveals distinct growth patterns. Front. Cell. Infect. Microbiol. 2014, 4, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Wang, L.; Hou, Y.; Yuan, H.; Chen, H. The role of tryptophan in Chlamydia trachomatis persistence. Front. Cell Infect. Microbiol. 2022, 12, 931653. [Google Scholar] [CrossRef]
  107. Borel, N.; Pospischil, A.; Hudson, A.P.; Rupp, J.; Schoborg, R.V. The role of viable but non-infectious developmental forms in chlamydial biology. Front. Cell. Infect. Microbiol. 2014, 4, 97. [Google Scholar] [CrossRef] [Green Version]
  108. Abu-Lubad, M.; Meyer, T.F.; Al-Zeer, M.A. Chlamydia trachomatis inhibits inducible NO synthase in human mesenchymal stem cells by stimulating polyamine synthesis. J. Immunol. (Baltim. Md. 1950) 2014, 193, 2941–2951. [Google Scholar] [CrossRef]
  109. Ziklo, N.; Huston, W.M.; Taing, K.; Katouli, M.; Timms, P. In vitro rescue of genital strains of Chlamydia trachomatis from interferon-γ and tryptophan depletion with indole-positive, but not indole-negative Prevotella spp. BMC Microbiol. 2016, 16, 286. [Google Scholar] [CrossRef] [Green Version]
  110. Molenaar, M.C.; Singer, M.; Ouburg, S. The two-sided role of the vaginal microbiome in Chlamydia trachomatis and Mycoplasma genitalium pathogenesis. J. Reprod. Immunol. 2018, 130, 11–17. [Google Scholar] [CrossRef]
  111. Singh, S.; Verma, N.; Taneja, N. The human gut resistome: Current concepts & future prospects. Indian J. Med. Res. 2019, 150, 345–358. [Google Scholar] [CrossRef]
  112. Lanza, V.F.; Baquero, F.; Martínez, J.L.; Ramos-Ruíz, R.; González-Zorn, B.; Andremont, A.; Sánchez-Valenzuela, A.; Ehrlich, S.D.; Kennedy, S.; Ruppé, E.; et al. In-depth resistome analysis by targeted metagenomics. Microbiome 2018, 6, 11. [Google Scholar] [CrossRef] [PubMed]
  113. Ballini, A.; Santacroce, L.; Cantore, S.; Bottalico, L.; Dipalma, G.; Vito, D.; Saini, R.; Inchingolo, F. Probiotics Improve Urogenital Health in Women. Open Access Maced. J. Med. Sci. 2018, 6, 1845–1850. [Google Scholar] [CrossRef] [Green Version]
  114. Kaur, H.; Ali, S.A. Probiotics and gut microbiota: Mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct. 2022, 13, 7423–7447. [Google Scholar] [CrossRef]
  115. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  116. Yoshikata, R.; Yamaguchi, M.; Mase, Y.; Tatsuyuki, A.; Myint, K.Z.Y.; Ohta, H. Evaluation of the efficacy of Lactobacillus-containing feminine hygiene products on vaginal microbiome and genitourinary symptoms in pre- and postmenopausal women: A pilot randomized controlled trial. PLoS ONE 2022, 17, e0270242. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, S.; Hugerth, L.W.; Schuppe-Koistinen, I.; Du, J. The right bug in the right place: Opportunities for bacterial vaginosis treatment. NPJ Biofilms Microbiomes 2022, 8, 34. [Google Scholar] [CrossRef]
  118. De Gregorio, P.R.; Juárez Tomás, M.S.; Nader-Macías, M.E. Immunomodulation of Lactobacillus reuteri CRL1324 on Group B Streptococcus Vaginal Colonization in a Murine Experimental Model. Am. J. Reprod. Immunol. 2016, 75, 23–35. [Google Scholar] [CrossRef] [PubMed]
  119. De Gregorio, P.R.; Maldonado, N.C.; Pingitore, E.V.; Terraf, M.C.L.; Tomás, M.S.J.; de Ruiz, C.S.; Santos, V.; Wiese, B.; Bru, E.; Paiz, M.C.; et al. Intravaginal administration of gelatine capsules containing freeze-dried autochthonous lactobacilli: A double-blind, randomised clinical trial of safety. Benef. Microbes 2020, 11, 5–17. [Google Scholar] [CrossRef] [PubMed]
  120. Kariyama, R.; Nasu, Y.; Ishii, A. Efficacy of Lactobacillus vaginal suppositories for the prevention of recurrent cystitis: A phase II clinical trial. Int. J. Urol. 2021, 28, 1026–1031. [Google Scholar] [CrossRef]
  121. Vladareanu, R.; Mihu, D.; Mitran, M.; Mehedintu, C.; Boiangiu, A.; Manolache, M.; Vladareanu, S. New evidence on oral L. plantarum P17630 product in women with history of recurrent vulvovaginal candidiasis (RVVC): A randomized double-blind placebo-controlled study. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 262–267. [Google Scholar] [CrossRef] [PubMed]
  122. Verdenelli, M.C.; Cecchini, C.; Coman, M.M.; Silvi, S.; Orpianesi, C.; Coata, G.; Cresci, A.; Di Renzo, G.C. Impact of Probiotic SYNBIO(®) Administered by Vaginal Suppositories in Promoting Vaginal Health of Apparently Healthy Women. Curr. Microbiol. 2016, 73, 483–490. [Google Scholar] [CrossRef] [PubMed]
  123. Mei, Z.; Li, D. The role of probiotics in vaginal health. Front. Cell. Infect. Microbiol. 2022, 12, 963868. [Google Scholar] [CrossRef] [PubMed]
  124. Cohen, C.R.; Wierzbicki, M.R.; French, A.L.; Morris, S.; Newmann, S.; Reno, H.; Green, L.; Miller, S.; Powell, J.; Parks, T.; et al. Randomized Trial of Lactin-V to Prevent Recurrence of Bacterial Vaginosis. N. Engl. J. Med. 2020, 382, 1906–1915. [Google Scholar] [CrossRef] [PubMed]
  125. Tomusiak, A.; Strus, M.; Heczko, P.B.; Adamski, P.; Stefański, G.; Mikołajczyk-Cichońska, A.; Suda-Szczurek, M. Efficacy and safety of a vaginal medicinal product containing three strains of probiotic bacteria: A multicenter, randomized, double-blind, and placebo-controlled trial. Drug Des. Devel. Ther. 2015, 9, 5345–5354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Heczko, P.B.; Tomusiak, A.; Adamski, P.; Jakimiuk, A.J.; Stefański, G.; Mikołajczyk-Cichońska, A.; Suda-Szczurek, M.; Strus, M. Supplementation of standard antibiotic therapy with oral probiotics for bacterial vaginosis and aerobic vaginitis: A randomised, double-blind, placebo-controlled trial. BMC Women’s Health 2015, 15, 115. [Google Scholar] [CrossRef] [Green Version]
  127. Pendharkar, S.; Brandsborg, E.; Hammarström, L.; Marcotte, H.; Larsson, P.G. Vaginal colonisation by probiotic lactobacilli and clinical outcome in women conventionally treated for bacterial vaginosis and yeast infection. BMC Infect. Dis. 2015, 15, 255. [Google Scholar] [CrossRef] [Green Version]
  128. Harasim-Dylak, A.; Roguska, M.; Maździarz, A. Role of Trivagin in restoration and maintenance of normal vaginal ecosystem in women treated for recurrent bacterial vaginosis. Curr. Gynecol. Oncol. 2011, 9, 245–252. [Google Scholar]
  129. Larsson, P.G.; Stray-Pedersen, B.; Ryttig, K.R.; Larsen, S. Human lactobacilli as supplementation of clindamycin to patients with bacterial vaginosis reduce the recurrence rate; a 6-month, double-blind, randomized, placebo-controlled study. BMC Women’s Health 2008, 8, 3. [Google Scholar] [CrossRef]
  130. Barbonetti, A.; Cinque, B.; Vassallo, M.R.; Mineo, S.; Francavilla, S.; Cifone, M.G.; Francavilla, F. Effect of vaginal probiotic lactobacilli on in vitro-induced sperm lipid peroxidation and its impact on sperm motility and viability. Fertil. Steril. 2011, 95, 2485–2488. [Google Scholar] [CrossRef]
  131. Mastromarino, P.; Hemalatha, R.; Barbonetti, A.; Cinque, B.; Cifone, M.G.; Tammaro, F.; Francavilla, F. Biological control of vaginosis to improve reproductive health. Indian J. Med. Res. 2014, 140 (Suppl. 1), S91–S97. [Google Scholar]
  132. Wang, J.; Si, W.; Du, Z.; Zhang, J.; Xue, M. Antioxidants in Animal Feed. Antioxidants 2022, 11, 1760. [Google Scholar] [CrossRef]
  133. Homayouni, A.; Bastani, P.; Ziyadi, S.; Mohammad-Alizadeh-Charandabi, S.; Ghalibaf, M.; Mortazavian, A.M.; Mehrabany, E.V. Effects of probiotics on the recurrence of bacterial vaginosis: A review. J. Low. Genit. Tract Dis. 2014, 18, 79–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Magrone, T.; Jirillo, E. The New Era of Nutraceuticals: Beneficial Effects of Polyphenols in Various Experimental and Clinical Settings. Curr. Pharm. Des. 2018, 24, 5229–5231. [Google Scholar] [CrossRef]
  135. Topi, S.; Bottalico, L.; Charitos, I.A.; Colella, M.; Di Domenico, M.; Palmirotta, R.; Santacroce, L. Biomolecular Mechanisms of Autoimmune Diseases and Their Relationship with the Resident Microbiota: Friend or Foe? Pathophysiology 2022, 29, 507–536. [Google Scholar] [CrossRef]
  136. Li, R.; Maimai, T.; Yao, H.; Liu, X.; He, Z.; Xiao, C.; Wang, Y.; Xie, G. Protective effects of polydatin on LPS-induced endometritis in mice. Microb. Pathog. 2019, 137, 103720. [Google Scholar] [CrossRef] [PubMed]
  137. Jøraholmen, M.W.; Škalko-Basnet, N.; Acharya, G.; Basnet, P. Resveratrol-loaded liposomes for topical treatment of the vaginal inflammation and infections. Eur. J. Pharm. Sci. 2015, 79, 112–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Caccavo, D.; Afeltra, A.; Pece, S.; Giuliani, G.; Freudenberg, M.; Galanos, C.; Jirillo, E. Lactoferrin-lipid A-lipopolysaccharide interaction: Inhibition by anti-human lactoferrin monoclonal antibody AGM 10.14. Infect. Immun. 1999, 67, 4668–4672. [Google Scholar] [CrossRef] [Green Version]
  139. Alexander, D.B.; Iigo, M.; Yamauchi, K.; Suzui, M.; Tsuda, H. Lactoferrin: An alternative view of its role in human biological fluids. Biochem. Cell Biol. 2012, 90, 279–306. [Google Scholar] [CrossRef]
  140. Valenti, P.; Rosa, L.; Capobianco, D.; Lepanto, M.S.; Schiavi, E.; Cutone, A.; Paesano, R.; Mastromarino, P. Role of Lactobacilli and Lactoferrin in the Mucosal Cervicovaginal Defense. Front. Immunol. 2018, 9, 376. [Google Scholar] [CrossRef] [Green Version]
  141. Kowalczyk, P.; Kaczyńska, K.; Kleczkowska, P.; Bukowska-Ośko, I.; Kramkowski, K.; Sulejczak, D. The Lactoferrin Phenomenon-A Miracle Molecule. Molecules 2022, 27, 2941. [Google Scholar] [CrossRef] [PubMed]
  142. Novak, R.M.; Donoval, B.A.; Graham, P.J.; Boksa, L.A.; Spear, G.; Hershow, R.C.; Chen, H.Y.; Landay, A. Cervicovaginal levels of lactoferrin, secretory leukocyte protease inhibitor, and RANTES and the effects of coexisting vaginoses in human immunodeficiency virus (HIV)-seronegative women with a high risk of heterosexual acquisition of HIV infection. Clin. Vaccine Immunol. CVI 2007, 14, 1102–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Sessa, R.; Di Pietro, M.; Schiavoni, G.; Petrucca, A.; Cipriani, P.; Zagaglia, C.; Nicoletti, M.; Santino, I.; del Piano, M. Measurement of Chlamydia pneumoniae bacterial load in peripheral blood mononuclear cells may be helpful to assess the state of chlamydial infection in patients with carotid atherosclerotic disease. Atherosclerosis 2007, 195, e224–e230. [Google Scholar] [CrossRef] [PubMed]
  144. Russo, R.; Karadja, E.; De Seta, F. Evidence-based mixture containing Lactobacillus strains and lactoferrin to prevent recurrent bacterial vaginosis: A double blind, placebo controlled, randomised clinical trial. Benef. Microbes 2019, 10, 19–26. [Google Scholar] [CrossRef] [PubMed]
  145. Miranda, M.; Saccone, G.; Ammendola, A.; Salzano, E.; Iannicelli, M.; De Rosa, R.; Nazzaro, G.; Locci, M. Vaginal lactoferrin in prevention of preterm birth in women with bacterial vaginosis. J. Matern.-Fetal Neonatal Med. 2021, 34, 3704–3708. [Google Scholar] [CrossRef] [PubMed]
  146. Trentini, A.; Maritati, M.; Rosta, V.; Cervellati, C.; Manfrinato, M.C.; Hanau, S.; Greco, P.; Bonaccorsi, G.; Bellini, T.; Contini, C. Vaginal Lactoferrin Administration Decreases Oxidative Stress in the Amniotic Fluid of Pregnant Women: An Open-Label Randomized Pilot Study. Front. Med. 2020, 7, 555. [Google Scholar] [CrossRef] [PubMed]
  147. Artym, J.; Zimecki, M. Antimicrobial and Prebiotic Activity of Lactoferrin in the Female Reproductive Tract: A Comprehensive Review. Biomedicines 2021, 9, 1940. [Google Scholar] [CrossRef]
  148. Yamauchi, K.; Wakabayashi, H.; Shin, K.; Takase, M. Bovine lactoferrin: Benefits and mechanism of action against infections. Biochem. Cell Biol. 2006, 84, 291–296. [Google Scholar] [CrossRef]
Figure 1. Local immunity in the FGT. Natural immunity and adaptive immunity contribute to the protection of the mucosal FGT, and also to maintaining a tolerogenic profile of the genital habitat.
Figure 1. Local immunity in the FGT. Natural immunity and adaptive immunity contribute to the protection of the mucosal FGT, and also to maintaining a tolerogenic profile of the genital habitat.
Life 13 01531 g001
Figure 2. Composition and functions of the vaginal microbiota. Lactobacillus spp. and their metabolites are able to increase the protective epithelial barrier, also acting as microbicidal and anti-inflammatory agents. pH < 4.5; L. a. concentration 120 mm; SCFAs concentration: acetate <120 mm.
Figure 2. Composition and functions of the vaginal microbiota. Lactobacillus spp. and their metabolites are able to increase the protective epithelial barrier, also acting as microbicidal and anti-inflammatory agents. pH < 4.5; L. a. concentration 120 mm; SCFAs concentration: acetate <120 mm.
Life 13 01531 g002
Table 1. Pathogenetic mechanisms responsible for vaginal infections. The bacteria responsible for infection perform different activities, aimed at decreasing the vaginal antimicrobial function, or utilizing substances necessary for these microbes’ growth.
Table 1. Pathogenetic mechanisms responsible for vaginal infections. The bacteria responsible for infection perform different activities, aimed at decreasing the vaginal antimicrobial function, or utilizing substances necessary for these microbes’ growth.
DiseasePathogenetic Mechanisms
Aerobic vaginitis
  • Increased production of pro-Inflammatory cytokines
  • Production of oxide-inducible lactate dehydrogenase with survival to lactic acid fermentation (S. aureus)
Bacterial vaginosis
  • Reduced release of lactic acid and tyrosine
  • Increased release of succinic acid with inhibition of leukocyte chemotaxis
Chlamydia trachomatis (C. t.) infection
  • Less production of lactic acid
  • Reduced vaginal levels of valine, tyramine, cadaverine and succinate
  • Utilization of tryptophan for its growth
  • Abrogation of anti-microbial activity of nitric oxide
Table 2. Main natural products used in the correction of FGT dysbiosis. Probiotics, lactoferrin, and polyphenols attempt to reconstitute the normal microbiota, and also conduct anti-inflammatory, antimicrobial, and immunomodulating activities at the vaginal level.
Table 2. Main natural products used in the correction of FGT dysbiosis. Probiotics, lactoferrin, and polyphenols attempt to reconstitute the normal microbiota, and also conduct anti-inflammatory, antimicrobial, and immunomodulating activities at the vaginal level.
ProbioticsLactoferrinPolyphenols
  • Production of bacteriocins
  • Adherence to epithelial cells
  • Generation of mucus components
  • Extracellular matrix-colonization
  • Modulation of the immune system
  • Increased levels of polydatin-mediated inhibition of lactoferrin in cervicovaginal fluid in FGT infections
  • Reduction of pathogens and inflammatory biomarkers in FGT dysbiosis, following oral vaginal administration
  • Polydatin-mediated inhibition of NF-kB pathway in murine endometritis
  • Resveratrol-loaded liposomes in vaginal inflammation
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

Santacroce, L.; Palmirotta, R.; Bottalico, L.; Charitos, I.A.; Colella, M.; Topi, S.; Jirillo, E. Crosstalk between the Resident Microbiota and the Immune Cells Regulates Female Genital Tract Health. Life 2023, 13, 1531. https://doi.org/10.3390/life13071531

AMA Style

Santacroce L, Palmirotta R, Bottalico L, Charitos IA, Colella M, Topi S, Jirillo E. Crosstalk between the Resident Microbiota and the Immune Cells Regulates Female Genital Tract Health. Life. 2023; 13(7):1531. https://doi.org/10.3390/life13071531

Chicago/Turabian Style

Santacroce, Luigi, Raffaele Palmirotta, Lucrezia Bottalico, Ioannis Alexandros Charitos, Marica Colella, Skender Topi, and Emilio Jirillo. 2023. "Crosstalk between the Resident Microbiota and the Immune Cells Regulates Female Genital Tract Health" Life 13, no. 7: 1531. https://doi.org/10.3390/life13071531

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