ArticlePDF AvailableLiterature Review

The Interaction Between Microorganisms, Metabolites, and Immune System in the Female Genital Tract Microenvironment

Frontiers
Frontiers in Cellular and Infection Microbiology
Authors:

Abstract and Figures

The female reproductive tract microenvironment includes microorganisms, metabolites, and immune components, and the balance of the interactions among them plays an important role in maintaining female reproductive tract homeostasis and health. When any one of the reproductive tract microorganisms, metabolites, or immunity is out of balance, it will affect the other two, leading to the occurrence and development of diseases and the appearance of corresponding symptoms and signs, such as infertility, miscarriage, premature delivery, and gynecological tumors caused by infectious diseases of the reproductive tract. Nutrients in the female reproductive tract provide symbiotic and pathogenic microorganisms with a source of nutrients for their own reproduction and utilization. At the same time, this interaction with the host forms a variety of metabolites. Changes in metabolites in the host reproductive tract are related not only to the interaction between the host and microbiota under dysbiosis but also to changes in host immunity or the environment, all of which will participate in the pathogenesis of diseases and lead to disease-related phenotypes. Microorganisms and their metabolites can also interact with host immunity, activate host immunity, and change the host immune status and are closely related to persistent genital pathogen infections, aggravation of infectious diseases, severe pregnancy outcomes, and even gynecological cancers. Therefore, studying the interaction between microorganisms, metabolites, and immunity in the reproductive tract cannot only reveal the pathogenic mechanisms that lead to inflammation of the reproductive tract, adverse pregnancy outcomes and tumorigenesis but also provide a basis for further research on the diagnosis and treatment of targets.
Content may be subject to copyright.
The Interaction Between
Microorganisms, Metabolites, and
Immune System in the Female
Genital Tract Microenvironment
Huanrong Li
1,2
, Yuqin Zang
1,2
, Chen Wang
1,2
, Huiyang Li
1,2
, Aiping Fan
1,2
, Cha Han
1,2
and Fengxia Xue
1,2
*
1
Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China,
2
Department of
Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University
General Hospital, Tianjin, China
The female reproductive tract microenvironment includes microorganisms, metabolites, and
immune components, and the balance of the interactions among them plays an important role
in maintaining female reproductive tract homeostasis and health. When any one of the
reproductive tract microorganisms, metabolites, or immunity is out of balance, it will affect the
other two, leading to the occurrence and development of diseases and the appearance of
corresponding symptoms and signs, such as infertility, miscarriage, premature delivery, and
gynecological tumors caused by infectious diseases of the reproductive tract. Nutrients in the
female reproductive tract provide symbiotic and pathogenic microorganisms with a source of
nutrients for their own reproduction and utilization. At the same time, this interaction with the
host forms a variety of metabolites. Changes in metabolites in the host reproductive tract are
related not only to the interaction between the host and microbiota under dysbiosis but also to
changes in host immunity or the environment, all of which will participate in the pathogenesis
of diseases and lead to disease-related phenotypes. Microorganisms and their metabolites
can also interact with host immunity, activate host immunity, and change the host immune
status and are closely related to persistent genital pathogen infections, aggravation of
infectious diseases, severe pregnancy outcomes, and even gynecological cancers.
Therefore, studying the interaction between microorganisms, metabolites, and immunity in
the reproductive tract cannot only reveal the pathogenic mechanisms that lead to
inammation of the reproductive tract, adverse pregnancy outcomes and tumorigenesis
but also provide a basis for further research on the diagnosis and treatment of targets.
Keywords: microenvironment, female genital tract, immunity, metabolites, microbiota
INTRODUCTION
Different from the high diversity of the gastrointestinal tract, the female genital tract microbiome has
low diversity, and it changes dynamically through the female menstrual cycle (Consortium, 2012;
Chen et al., 2017). Most microbes have a symbiotic relationship with the host. Accounting for 9095%
of the total bacterial biomass, Lactobacillus spp. represents a healthy female genital tract microbiota
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094881
Edited by:
Carla R. Taddei,
University of São Paulo, Brazil
Reviewed by:
Silvia Daher,
Federal University of São Paulo, Brazil
Alison Jane Carey,
Queensland University of Technology,
Australia
*Correspondence:
Fengxia Xue
fengxiaxue1962@gmail.com
These authors have contributed
equally to this work
Specialty section:
This article was submitted to
Microbiome in Health and Disease,
a section of the journal
Frontiers in Cellular
and Infection Microbiology
Received: 23 September 2020
Accepted: 18 November 2020
Published: 23 December 2020
Citation:
Li H, Zang Y, Wang C, Li H, Fan A,
Han C and Xue F (2020) The
Interaction Between Microorganisms,
Metabolites, and Immune System in
the Female Genital Tract
Microenvironment.
Front. Cell. Infect. Microbiol. 10:609488.
doi: 10.3389/fcimb.2020.609488
REVIEW
published: 23 December 2020
doi: 10.3389/fcimb.2020.609488
that produces lactic acid to maintain an acidic microenvironment.
It can also inhibit pathogens through competition, adhesion
prevention, and the secretion of antibacterial and
immunomodulatory substances (Anahtar et al., 2018;Van der
Veer et al., 2019). Vaginitis, cervicitis, and pelvic inammatory
disease (PID) will occur if pathogenic bacteria surpass lactobacilli
in the female genital tract and can cause uncomfortable
symptoms such as increased vulvovaginal discharge, itching,
odor, and lower abdominal pain (Workowski and Bolan, 2015).
However, there are differences in the microbes between
subjects and in the ability of the host to resist dysbiosis that
may be related to race, diet, age, living habits, immunity, disease
susceptibility, and genetic polymorphism (Consortium,
2012;Anahtar et al., 2018;Chu et al., 2018;Serrano et al.,
2019). Furthermore, the dominance of different microora is not
necessarily related to symptoms because partial non-Lactobacillus-
dominant women do not experience uncomfortable symptoms of
vulvovaginitis; hence, we cannot dene disease by the number of
bacteria alone, and we cannot dene dysbiosis without the internal
milieu of the host and disease environment (Anahtar et al., 2018;
Scott et al., 2019).
Metabolites in the reproductive tract play an important role
in female genital tract inammation, pregnancy and tumors and
can be considered biomarkers of disease severity, diagnosis, and
prognosis (Ghartey et al., 2015;McMillan et al., 2015;Ilhan et al.,
2019;Song et al., 2019). Metabolites in the female reproductive tract
are the substrates, intermediates and byproducts of biochemical
reactions caused by the interaction of human nutrients and bacteria,
reecting downstream events of gene expression (Altmäe et al.,
2014;McMillan et al., 2015;Turkoglu et al., 2016;Watson and Reid,
2018)(Figure 1). These metabolites are better than genome,
transcriptome, and proteome substances at predicting the disease
phenotype (Altmäe et al., 2014). Genital infections, adverse
pregnancy outcomes, and cancers possess different metabolic
signatures that are often accompanied by dysbiosis of the female
genital tract (Ghartey et al., 2015;Ceccarani et al., 2019;Ilhan et al.,
2019). The metabolic pathways affected by these metabolites mainly
include amino acids, carbohydrates, and lipid metabolism, which
are closely related to life activities (Srinivasan et al., 2015). These
activities further affect host cell function, immunity, and disease
susceptibility and help maintain the balance of the hosts
reproductive tract microenvironment.
The hosts innate and adaptive immune systems perform
complex interactions with microorganisms and metabolites
(Agostinis et al., 2019;Delgado-Diaz et al., 2019). Microbial
ligands bind to host receptors to produce inammatory factors,
chemokines and antimicrobial products to regulate the immune
response of the reproductive tract (Hooper et al., 2012).Vaginal
dysbiosis cannot only directly cause vaginal epithelial injury
through pathogens (Tao et al., 2019), but also indirectly cause
vaginal epithelial injury through immune components, which in
turn release metabolites into the microenvironment (Olive and
Sassetti, 2016;Serrano et al., 2019). This metabolite may be
ingested by the vaginal microbiota, leading to increased
microbial metabolism, which is benecial to the growth and
reproduction of the microbiota (Serrano et al., 2019). The local
competition between the host, pathogen and different immune
cells for metabolic precursors will also affect the ability of immune
cells to respond effectively to infection, affecting the growth and
immunogenicity of the pathogen and further affecting the host
response (Hooper et al., 2012;Olive and Sassetti, 2016;Postler and
Ghosh, 2017). Therefore, the interaction between microorganisms,
metabolites, and immunity in the host reproductive tract
microenvironment plays an important role in maintaining the
balance of the reproductive tract (Pruski et al., 2018). An
imbalance in any part will result in host phenotype changes,
disease, and even serious complications. Therefore, this article
intended to review the relationship and importance of
FIGURE 1 | Microenvironmental disorders of the female reproductive tract are closely related to inammations, adverse pregnancy outcomes, and tumors. Modied
from PawełŁaniewski et al. (2020).
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094882
reproductive tract microorganisms, metabolites, and immunity to
obtain a deeper understanding of the reproductive tract
microenvironment, reproductive tract diseases and adverse
reproductive tract outcomes.
NORMAL VAGINAL MICROENVIRONMENT
The vaginal microbiota community state types (CSTs) of women
of childbearing age are divided into ve categories (Ravel et al.,
2011). CST I is dominated by Lactobacillus crispatus; CST II by L.
gasseri; CST V by L. jensenii; and CST III by L. iners. CST IV
belongs to the Lactobacillus-decient type, which is dominated
by anaerobic bacteria (classied by bacterial vaginosis, BV),
partial aerobic bacteria (classied by aerobic vaginitis, AV) or a
modest proportion of Lactobacillus spp. (Gajer et al., 2012). The
vaginal microbiota is dynamic and occasionally transitions to an
intermediate state or a disease state in most normal non-
pregnant women. A high Nugent score does not indicate the
disease status or microecological disorders (Gajer et al., 2012).
The internal milieu of the host will make the microbiota return to
a stable state. Factors that cause vaginal microbiota changes are
primarily related to menstruation. Others include sexual
intercourse, hormonal contraception, antimicrobial agents, use
of lubricants, and vaginal douching, but they have less impact
than menstrual periods and cause changes in the vaginal
microbiota for a shorter duration (Gajer et al., 2012;Mitchell
et al., 2012). In addition, in a few women, the vaginal CST does
not change with the menstrual cycle and hormonal
contraception, but further study by future researchers is
needed to determine whether it is related to the metabolic
function of bacteria (Gajer et al., 2012;Song et al., 2020).
Lactobacillus abundance in the female genital tract is strongly
positively correlated with lactate and 4-hydroxyphenylacetate
and correlated to a lesser extent with isoleucine, leucine,
tryptophan, phenylalanine, aspartate, dimethylamine, sarcosine
and pi-methylhistidine, all of which are typically associated with
vaginal health (Srinivasan et al., 2015;Ceccarani et al., 2019)
(Table 1). L. crispatus and L. jensenii have similar metabolic
patterns (Srinivasan et al., 2015), while, L. crispatus and L. iners
have different metabolic characteristics (Pruski et al., 2018). For
example, studies have found that the genome of L. crispatus is
almost twice that of L. iners (France et al., 2016). However, the
carbon metabolism of L. iners is fermented by fewer compounds
than L. crispatus (Pruski et al., 2018). When the dominant
bacteria are L. crispatus and/or L. jensenii,mostofthe
metabolites in the vagina are amino acids and dipeptide, such
as higher levels of ornithine, lysine, glycylproline, phenylalanine
(Srinivasan et al., 2015). Similar to BV-related ora, L. iners are
correlated with amino acid catabolites, such as higher levels of
TABLE 1 | Current existing articles analyzing the correlation between the genital tract ora and metabolites in normal non-pregnant women of reproductive age.
Year Author Population Sample CST Metabolites
2019 (Ceccarani
et al.,
2019)
Healthy women (n = 21), women
with BV (n = 20) and women with
Chlamydia trachomatis infection
(n = 20)
Vaginal
swabs
Contains all
Lactobacillus spp.
without
distinguishing CST
High levels of lactate, 4-hydroxyphenylacetate, isoleucine, leucine, tryptophan,
phenylalanine, aspartate, dimethylamine, sarcosine and pi-methylhistidine
2018 (Parolin
et al.,
2018)
Healthy women (n = 22), women
with Chlamydia trachomatis
infection (n = 20), and women with
BV (n = 19)
Vaginal
swabs
Contains all
Lactobacillus spp.
without
distinguishing CST
Higher levels of lactate, 4-hydroxyphenylacetate, diverse amino acids
(phenylalanine, glutamate, leucine, threonine, tryptophan, aspartate) and
amino acid derivatives(sarcosine)
2015 (Srinivasan
et al.,
2015)
Women with BV (n = 40) and
women without BV (n = 20)
Vaginal
uid
CST-I 1) Higher levels of sugars (maltose, maltotriose, and maltohexose), lipid
metabolism biochemicals (such as arachidonate and carnitine), amino acids
(ornithine, lysine), dipeptide (glycylproline, phenylalanine) as well as lactate,
and urea
2) Lower levels of N-acetylneuraminate, succinate, the carnitine precursor
deoxycarnitine, the eicosanoid 12-hydroxyeicosatetraenoic acid, the fatty acid
13-hydroxyoctadecadienoic acid, the nucleobase uracil, and glutathione
CST-III 1) Higher levels of proline, threonine, aspartate, serine, and valinylglutamate
2) Lower levels of glutamate and glycylleucine
CST-V Same as CST-I:
1) Higher levels of sugars (maltose, maltotriose, and maltohexose), lipid
metabolism biochemicals (such as arachidonate and carnitine), amino acids
(ornithine, lysine), dipeptide (glycylproline, phenylalanine) as well as lactate,
and urea
2) Lower levels of N-acetylneuraminate, succinate, the carnitine precursor
deoxycarnitine, the eicosanoid 12-hydroxyeicosatetraenoic acid, the fatty acid
13-hydroxyoctadecadienoic acid, the nucleobase uracil, and glutathione
2015 (McMillan
et al.,
2015)
Pregnant women (n = 67) and non-
pregnant women (n = 64)
Vaginal
uid
CST-I Higher levels of succinate
2012 (Gajer
et al.,
2012)
Reproductive age women (n = 32) Vaginal
swabs
CST-III 1) Higher levels of lactate
2) Lower levels of succinate and acetate
CST, Community state types; BV, bacterial vaginosis.
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094883
proline, threonine, aspartate, serine, and valinylglutamate
(Srinivasan et al., 2015). In addition, L. iners often has a
symbiotic relationship with G. vaginalis, and both produce
similar levels of cholesterol-dependent cytolysin (Macklaim
et al., 2013). Therefore, the metabolic characteristics of L.
crispatus and/or L. jensenii dominance can be dened as a
healthy vaginal microenvironment. However, whether the
metabolic characteristics of the non-Lactobacillus abundance of
some asymptomatic women are similar to those of L. crispatus
and/or L. jensenii dominance needs to be further explored.
The mucous layer on the surface of female genital tract
epithelial cells plays an important role as the rst line of
defense against microbial invasion (Mirmonsef et al., 2011;
Aldunate et al., 2015). When microorganisms break through
the line of defense, epithelial cells use pattern recognition
receptors (PPRs) to identify microorganisms to produce
inammatory factors and recruit inammatory cells to resist
microbial invasion and colonization. The dominance of
Lactobacillus in the genital tract is essential, as it inhibits
pathogens and maintains immune equilibrium (Smith and
Ravel, 2017). Studies have found that the concentration of
inammatory factors in the vagina is very low when L.
crispatus and L. jensenii are dominant (Kyongo et al., 2012).
Lactic acid, as a metabolite derived primarily from Lactobacillus
spp., is also related to reproductive tract immunity (Delgado-
Diaz et al., 2019). L-lactic acid produced by Lactobacillus spp. can
cause an anti-inammatory response and inhibit the production
of proinammatory cytokines and chemokines induced by toll-
like receptor (TLR) in cervical and vaginal epithelial cells at low
pH (Delgado-Diaz et al., 2019). In addition, lactic acid can
induce the secretion of the anti-inammatory cytokine
interleukin (IL)-10, reduce the production of the
proinammatory cytokine IL-12 in dendritic cells (DCs), and
reduce the cytotoxicity of natural killer cells (Ilhan et al., 2019).
The anti-inammatory activity of lactic acid also requires the
presence of organic acids produced by microorganisms to
maintain vaginal health, mainly by increasing the production
of the anti-inammatory cytokine IL-1RA, inhibiting the
proinammatory signal of the IL-1 cytokine, and slightly
reducing the production of the proinammatory cytokines IL-6
and macrophage inammatory protein 3 alpha (MIP-3a)
(Delgado-Diaz et al., 2019). Therefore, the interaction between
the ora, metabolites, and immunity in the healthy reproductive
tract is very important for maintaining the health of the
reproductive tract. When any one party is imbalanced, it will
affect the balance of the reproductive tract.
COMMON REPRODUCTIVE TRACT
INFECTIONS
Female genital tract infections mainly include vaginitis, cervicitis,
and PID (Sherrard et al., 2018). The main cause is exogenous
pathogen interference or endogenous dysbiosis (Workowski and
Bolan, 2015;Song et al., 2020). At present, the relevant research
on infectious diseases caused by the interaction between
reproductive tract microorganisms, metabolites and the host is
mainly focused on BV, Chlamydia trachomatis (C. trachomatis),
and AV (Ceccarani et al., 2019). In addition, inammation of the
reproductive tract caused by bacterial ora disorders involving
BV, AV, and C. trachomatis infection is closely related to adverse
pregnancy outcomes and tumors (Sherrard et al., 2018). Other
diseases, such as trichomoniasis (Trichomonas vaginalis),
vulvovaginal candidiasis, and gonorrhea, have received less
relevant research in this area and may become future research
directions. Therefore, this section mainly discusses the
interaction between the microorganisms, metabolites, and host
immunity of three common RTIs: BV, C. trachomatis infection,
and AV.
BV
BV is the most common vaginal microbial disorder of women of
childbearing age, and can lead to adverse obstetrics and
gynecological outcomes such as infertility, miscarriage,
premature rupture of membranes, and premature delivery
(Workowski and Bolan, 2015;Baqui et al., 2019;Peebles et al.,
2019). It also increases the risk of sexually transmitted infections
(Shipitsyna et al., 2020). BV is characterized by an increase in the
diversity of vaginal microbiota, a decrease in Lactobacillus spp. in
the vagina, and an increase in BV-related anaerobic and
microaerobes (Srinivasan et al., 2015). BV-related bacteria
mainly include Gardnerella,Atopobium,Mycoplasma,
Megasphaera,Mobiluncus,Roseburia,Dialister.,Sneathia and
Prevotella spp. (McMillan et al., 2015;Ceccarani et al., 2019).
However, ora analysis alone cannot distinguish between a
normal vaginal environment and BV because Atopobium spp.,
Prevotella spp. and Mycoplasma hominis can also be detected in
healthy people, therefore, the vaginal microenvironment needs to
be analyzed in combination with metabolomics (Vitali
et al., 2015).
BV is closely related to metabolites in the genital tract
(Spiegel et al., 1980;Wolrath et al., 2002)(Table 2). The
metabolites of amines, organic acids, short chain fatty acids
(SCFAs), amino acids, nitrogenous bases and monosaccharides
of BV patients are signicantly different from those of
healthy individuals (Vitali et al., 2015). Current studies suggest
that metabolites better reect the disease phenotype than
microorganisms. Before disease symptoms appear, the
appearance or disappearance of certain metabolites in the
vagina has a positive or negative correlation with the metabolic
function of certain microorganisms (Yeoman et al., 2013).
Changes in maltose, kynurenine, nicotinate, malonate, acetate
and nicotinamide adenine dinucleotide (NAD
+
) represent the
occurrence of BV and can be used as metabolic biomarkers to
distinguish BV from a healthy vagina (Vitali et al., 2015). When
BV is cured, the metabolites associated with BV decrease
signicantly (Stanek et al., 1992;Srinivasan et al., 2015). In
addition, genital tract metabolic analysis plays a prominent
role in the diagnosis of BV (Watson and Reid, 2018). In 2015,
McMillan et al. (2015) foundthatanincreasein2-
hydroxyisovalerate and g-hydroxybutyrate and a decrease in
lactic acid and tyrosine in the vagina are the most sensitive
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094884
TABLE 2 | Current existing articles analyzing the correlation between the genital tract ora and metabolites in women with vaginitis.
Year Author Population Sample CST Metabolites
BV
2019 (Ceccarani
et al., 2019)
Healthy women (n = 21), women with BV (n = 20) and women
with Chlamydia trachomatis infection (n = 20)
Vaginal
swabs
CST-IV 1) Higher levels of organic acids (i.e.: formate, pyruvate,
propionate, acetate, 2-hydroxyisovalerate), amines (i.e.:
trimethylamine, putrescine), amino acids (i.e.: proline
and alanine) and 5-aminopentanoate
2) Lower levels of lactate, 4-hydroxyphenylacetate,
phenylalanine, pi-methylhistidine, glycine, isoleucine,
leucine, tryptophan, aspartate, dimethylamine, and
sarcosine
2018 (Parolin et al.,
2018)
Healthy women (n = 22), women with Chlamydia trachomatis
infection (n = 20), and women with BV (n = 19)
Vaginal
swabs
CST-IV Higher levels of biogenic amines (methylamine,
putrescine, trimethylamine, tyramine, desaminotyrosine),
organic acids (succinate, malonate, 2-
hydroxyisovalerate, and short-chain fatty acids) and
alanine
2015 (McMillan et al.,
2015)
Pregnant women (n = 67) and non-pregnant women (n = 64) Vaginal
uid
CST-IV 1) Organic acid: higher levels of 2-hydroxyisovalerate, g
-hydroxybutyrate, 2-hydroxyglutarate and 2-
hydroxyisocaproate; lower levels of lactate
2) Amines: higher levels of tyramine, putrescine, and
cadaverine
3) Amino acids: lower levels of tyrosine
2015 (Srinivasan
et al., 2015)
Women with BV (n = 40) and women with non-BV (n = 20) Vaginal
uid
CST-IV 1) Amino acid: higher levels of cadaverine, pipecolate,
tyramine, 4-hydroxyphenylacetate, 3- (4-hydroxyphenyl)
propionate, tryptamine, citrulline and putrescine; lower
concentrations of arginine, ornithine, spermine and
dipeptides
2) Carbohydrates: higher levels of N-acetylneuraminate,
galactose, threitol and succinate; lower levels of
glucosamine, maltotriose, maltotetraose,
maltopentaose, maltohexaose, lactate, fructose, and
mannitol
3) NAD: lower levels of nicotinamide; higher levels of
nicotinate
4) Lipids: higher levels of 12-hydroxyeicosatetraenoic
acid, deoxycarnitine, 4-hydroxybutyrate and
13-hydroxyoctadecadienoic acid; lower levels of
arachidonate, carnitine, ascorbic acid, acetylcarnitine,
propionylcarnitine, butyrylcarnitine, glycerol and
glycerol-3-phosphate
2015 (Vitali et al.,
2015)
BV-affected patients (n = 43) and healthy controls (n = 37) Vaginal
uid
CST-IV 1) Amines: higher levels of tyramine, ethanolamine,
trimethylamine, methylamine, cadaverine
2) Organic acids: higher levels of formate, malonate,
succinate, pyruvate, acetate
3) Short-chain fatty acids: higher levels of propionate,
butyrate, 2-hydroxyisovalerate
4) Amino acids: higher levels of proline; lower levels of
tryptophan, phenylalanine, tyrosine, glutamate,
isoleucine, leucine
5) Nitrogenous bases: higher levels of nicotinate, uracil;
lower levels of NAD+, inosine
6) Sugars: higher levels of glucose; lower levels of
maltose
7) Others: higher levels of urocanate, 2-aminoadipate,
3-methyl-2-oxovalerate; lower levels of kynurenine, sn-
glycero-3-phosphocholine, sarcosine
2013 (Yeoman et al.,
2013)
Pre-menopausal women of reproductive age (n = 36) Vaginal
lavage
uid
CST-IV 1) Higher levels of putrescine, cadaverine, 2-methyl-2-
hydroxybutanoic acid, hydroxylamine, glycolic acid,
tetradecanoic acid, and butyrolactone
2) Lower levels of 2,3-hydroxypropyl-2-aminoethyl
phosphate, cis-11-octadecanoic acid, and ribose-5-
phosphate
2012 (Gajer et al.,
2012)
Reproductive age women (n = 32) Vaginal
swabs
CST-IV 1) Higher levels of succinate and acetate
2) Lower concentrations of lactate
(Continued)
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094885
and specic indicators for the diagnosis of BV. Therefore, not
only the microbiota but also metabolites can be used as effective
reference indicators for clinical diagnosis.
The vaginal microbiota and metabolites of BV patients are also
closely related to the clinical symptoms and signs of the host. The
odor of vaginal secretions in patients with BV is related to the
increase in tyramine, trimethylamine, cadaverine, and putrescine
and the decrease in the aromatic substances 2 (5H)-furanone and
2-ethyl-4-methyl-1,3-dioxolane (Yeoman et al., 2013;Srinivasan
et al., 2015;Vitali et al., 2015). Odor is also closely related to
Dialister spp. (Yeoman et al., 2013;McMillan et al., 2015;
Srinivasan et al., 2015). Thin and homogeneous secretions are
positively related to cadaverine, and cadaverine is related to
Streptococcus spp. and Mycoplasma spp. (Yeoman et al., 2013;
Srinivasan et al., 2015). Clue cells are positively correlated
with deoxycarnitine and pipecolate, while deoxycarnitine is
positively correlated with BV-associated bacterium 1 (BVAB1),
Megasphaera sp. type 2, and several Prevotella species (Srinivasan
et al., 2015). Vaginal discharge is related to 2-methyl-2-
hydroxybutanoic acid and Mobiluncus spp. (Yeoman et al.,
2013). In addition, the metabolic pathways of amino acids,
carbohydrates, NAD, and lipids in the vaginal ora of BV
patients are active and are closely related to cellular life activities
(Srinivasan et al., 2015;Ceccarani et al., 2019).Therefore,
understanding the interaction between the ora and metabolites
of BV patients provides a basis for understanding the molecular
mechanisms of microbe-microbe and microbe-host interactions
(Ilhan et al., 2019).
BV-related bacteria can activate the hostsgenitaltract
immune response, but they do not cause obvious inammatory
symptoms such as redness, swelling, heat and pain (Smith and
Ravel, 2017). The reason may be related to the inuence of BV-
related microorganisms and their metabolites on immunity. In
2019, Delgado-Diaz et al. (2019) found that the sustained action
of organic acids, metabolites of the vaginal microbiota associated
with BV, led to dysregulation of the immune response of cervical
and vaginal epithelial cells in vitro. SCFAs can recruit and
activate female reproductive tract innate immune cells, such as
neutrophils and monocytes (Vitali et al., 2015). However, SCFAs
can also inhibit the production of proinammatory cytokines
and affect the migration and phagocytic response of immune
cells to regulate the immune response (Al-Mushrif et al., 2000).
In addition, succinic acid produced by Prevotella spp. and
Mobiluncus spp. in the genital tract can also inhibit leukocyte
chemotaxis and regulate the immune response (Al-Mushrif et al.,
2000;McMillan et al., 2015;Vitali et al., 2015). In 1985, Rotstein
et al. (1985) demonstrated that succinic acid has the strongest
chemotaxis inhibitory effect at pH 5.5 and at concentrations of
2030 mM. Therefore, the current research has proven that the
interaction between BV ora, metabolites and immunity is of
great signicance for understanding clinical symptoms and signs.
However, more research on the interaction mechanism between
immunity and metabolites in BV patients is needed to conrm
the inuence of metabolites on ora and immunity.
Chlamydia trachomatis
In 2016, the World Health Organization announced the newest
global C. trachomatis prevalence rate of 1.57% for women aged
1549 years and an estimated 127 million new cases women
worldwide that year (Organization, 2020). Most women infected
with C. trachomatis are asymptomatic (Ceccarani et al., 2019).
Approximately 10% of C. trachomatis infections will progress to
PID without timely treatment, which will cause severe ectopic
pregnancy, reproductive dysfunction and cancer (Workowski
and Bolan, 2015;Idahl et al., 2020). Lactic acid is an important
inhibitor of C. trachomatis infection (Gong et al., 2014).
However, L. iners produces less lactic acid, so the microbiota
dominated by L. iners increases the risk of C. trachomatis
infection (Van Houdt et al., 2018). Similarly, BV also increases
the risk of C. trachomatis infection due to a reduction in the
lactate concentration (Shipitsyna et al., 2020). Therefore, C.
trachomatis infection is greatly affected by lactic acid in the
reproductive tract microenvironment.
The interactions among microorganisms, C. trachomatis and
metabolites in the reproductive tract are closely related (Table 2).
In 2016, Ceccarani et al. (2019) performed a combined
metagenomic and metabolomics analysis on the vaginal
secretions of non-pregnant Caucasians of childbearing age with
risk factors for C. trachomatis infection. The study showed that C.
trachomatis infection was dominated by Lactobacillus in most
TABLE 2 | Continued
Year Author Population Sample CST Metabolites
2002 (Wolrath et al.,
2002)
Women of childbearing age with various lower genital tract
disorders (n = 61)
Vaginal
uid
CST-IV Higher levels of trimethylamine
1980 (Spiegel et al.,
1980)
Women with non-specic vaginitis (n = 53) Vaginal
uid
CST-IV 1) Higher levels of succinate, acetate, butyrate, and
propionate
2) Lower levels of lactate
Chlamydia trachomatis
2018 (Parolin et al.,
2018)
Women with Chlamydia trachomatis infection (n = 20), healthy
women (n = 22), and women with BV (n = 19)
Vaginal
swabs
CST-III Lower levels of tyramine, dimethylamine, cadaverine,
succinate, valine, isoleucine, glycine, sarcosine,
creatinine, 4-aminobutyrate
2019 (Ceccarani
et al., 2019)
Healthy women (n = 21), women with BV (n = 20) and women
with Chlamydia trachomatis infection (n = 20)
Vaginal
swabs
CST-IV Lower levels of lactate, certain amino acids and
biogenic amines
AV
2012 (Gajer et al.,
2012)
Reproductive age women (n = 32) Vaginal
swabs
CST-IV Higher levels of acetate and lactate
CST, Community state types; BV, bacterial vaginosis; AV, aerobic vaginitis.
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094886
people, and L. iners was increased, and some patients had
anaerobic bacteria as the dominant bacteria. Compared with
healthy controls, women infected with C. trachomatis showed
only slight changes in vaginal metabolites that were mainly
manifested as a reduction in certain amino acids and biogenic
amines (Ceccarani et al., 2019). In 2018, Parolin et al. (2018)
studied the characteristics of vaginal microbes and metabolites in
thecaseofC. trachomatis infection and found that vaginal valine,
isoleucine, tyramine, cadaverine, and succinate in patients with C.
trachomatis infection were signicantly decreased compared with
those in healthy controls, indicating that C. trachomatis may use
nitrogen as the rst nutrient source or that C. trachomatis may
affect the nitrogen metabolism of infected host cells. There is a
correlation between the vaginal microbiome, metabolites, and
genital symptoms of C. trachomatis infection (Parolin et al.,
2018). More than half of C. trachomatis-infected patients are
completely asymptomatic, while symptomatic patients mainly
manifest with abnormal vaginal discharge, dyspareunia, dysuria,
and abnormal bleeding. The concentration of 4-aminobutyrate is
signicantly different between asymptomatic and symptomatic
women with C. trachomatis infection. However, all asymptomatic
women have L. crispatus as the dominant bacteria in the vagina,
and only half of symptomatic women have L. crispatus as the
dominant bacteria (Parolin et al., 2018). Therefore, C. trachomatis
infection is related to the genital tract ora, metabolites and
clinical symptoms. However, the effect of 4-aminobutyrate on
host immunity against C. trachomatis infection needs
further study.
There are complicated interactions between C. trachomatis,
microorganisms, genital tract metabolites and immunity (Ziklo
et al., 2016b). Epithelial cells and immune cells infected by C.
trachomatis can secrete several proinammatory cytokines and
chemokines to eliminate pathogen infection (Rasmussen et al.,
1997;Johnson, 2004;Brunham and Rey-Ladino, 2005).
Interferon (IFN)-gis an important factor that inhibits the
reproduction of C. trachomatis (Shemer and Sarov, 1985). The
ability to synthesize tryptophan in the IFNg-rich infection
microenvironment is an important virulence factor of the
genital C. trachomatis serovars (Aiyar et al., 2014). IFN-g
mediates the activation of host indoleamine 2,3-dioxgenase
(IDO), leading to the consumption of tryptophan necessary for
the growth of C. trachomatis and inhibiting the growth of C.
trachomatis (Beatty et al., 1994;Aiyar et al., 2014;Olive and
Sassetti, 2016;Molenaar et al., 2018). The tryptophan needed for
the growth of C. trachomatis is reduced, and C. trachomatis
forms a static state (Byrne et al., 1989). It is known that BV
infection increases the risk of C. trachomatis infection. BV-
related bacteria, such as partial Prevotella species, can produce
indole (Romanik et al., 2007;Sasaki-Imamura et al., 2011), which
is increased in the vaginal discharge of BV patients (Lewis et al.,
2014). C. trachomatis in the genital tract can use indole produced
by microorganisms as a substrate to activate alternative
tryptophan synthesis pathways- the trpA,trpB and trpR genes,
synthesize tryptophan, and make C. trachomatis evade the
clearance of IFN-gin the genital tract (Fehlner-Gardiner et al.,
2002;Wood et al., 2003;Ziklo et al., 2016b). However, in patients
who are coinfection with BV and C. trachomatis, the inhibitory
response to IFN-gis not exactly the same, which may be related
to the level of indole in the vaginal microenvironment (Lewis
et al., 2014). In addition, the low oxygen environment formed
under BV may result in insufcient energy supply for the IFN-g
signaling pathway, which further reduces its function (Roth et al.,
2010). IFN-ginduces the production of nitric oxide and further
inhibits the growth of C. trachomatis (Agrawal et al., 2011). The
most recent in vitro experiments have conrmed that C.
trachomatis induces the expression of ornithine decarboxylase
(ODC), deprives the inducible nitric oxide synthase (iNOS)
substrate arginine, and actively promotes polyamine synthesis
while downregulating iNOS expression and inhibiting the
activity of iNOS to reduce nitric oxide production in the host
and further escape the hosts innate immunity (Abu-Lubad et al.,
2014;Olive and Sassetti, 2016). Studies have shown that the
amino acids and sugars in the environment are critical to the
ability of C. trachomatis to infect (Harper et al., 2000). However,
the detailed metabolic and immune interaction mechanisms still
need further study. After C. trachomatis escapes the hosts
immunity, the hosts immune surveillance is reduced, and the
environment is conducive to the growth of C. trachomatis, which
is reactivated (Belland et al., 2003). After C. trachomatis
reinfection or chronic infection, T helper (Th)1-, Th2- and
Th17-typecellsaretriggeredtomediatetissuedestruction,
brosis, and scarring, further leading to the progression of PID
and its sequelae (Ziklo et al., 2016a;Molenaar et al., 2018).
AV
The incidence of AV in women of childbearing age is approximately
10% (Donders et al., 2017). Signicantly different from BV patients
and those with a normal ora, AV patients have increased aerobic
bacteria or enterococci, such as Escherichia coli,Streptococcus
agalactiae,Staphylococcus aureus,Staphylococcus epidermidis,
Streptococcus anginosus,andEnterococcus faecalis, in the vagina
(Donders et al., 2017;Tao et al., 2019;Wang et al., 2020). Studies
have also shown that BV-related bacteria often appear in the vaginal
ora of AV patients, which may be related to the symbiotic
relationship between BV- and AV-related bacteria in the state of
ora disorder (Wang et al., 2020). Different from the clinical
symptoms and signs of BV, AV mainly manifests as foul and
yellow purulent discharge, but similar to BV, it easily causes adverse
obstetrics and gynecology complications that may be related to
bacterial ascending infection (Donders et al., 2017).
It is known that AV-related bacteria and their metabolites are
involved in the hostsinammatory state and immune response,
but their correlation with host disease phenotypes and diagnostic
applications have not yet been studied. Previous studies have
found that when Streptococcus sp. increase in the vaginal
secretions of non-pregnant women, acetate also increases
(Gajer et al., 2012)(Table 2). Acetate is a SCFAs that directly
activates the hostsimmune-inammatory pathway and
promotes the expansion of T-regulatory (Treg) cells (Olive and
Sassetti, 2016;Postler and Ghosh, 2017). However, the detailed
mechanism by which acetic acid is produced by Streptococcus sp.
and genital tract immunity still needs to be studied.
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094887
AV patients mainly present with a local immune imbalance in
the reproductive tract caused by pathogens (Benner et al., 2018).
In 2020, Budilovskaya et al. (2020) found that the expression of
IL1b, IL-6, IL-8, IL10, tumor necrosis factor-a(TNFa) and
CD68 messenger ribose nucleic acids (mRNAs) in AV patients
was signicantly increased, and this change was related to itching
or burning as well as increases in leukocytes and parabasal
epithelial cells under the microscope (Smith and Ravel, 2017).
Purulent vaginal discharge and vaginal redness may be related to
the toxic effect of the virulence gene sag of Streptococcus
anginosus on epithelial cells, leading to epithelial cell lysis (Tao
et al., 2019).
The molecular mechanism of inammatory genital tract
symptoms in AV patients may be related to the interaction of
metabolism and immunity. Nitric oxide plays an important role
in host resistance to pathogens. Nitric oxide is synthesized by
iNOS in inammatory cells (Richardson et al., 2006;Jones et al.,
2010). After the cells secrete nitric oxide, they can kill pathogens
directly beside the inammatory cells. However, Staphylococcus
aureus can evade the host nitric oxide response by changing
metabolism (Olive and Sassetti, 2016). Staphylococcus aureus
induces the expression of avohaemoglobin (Hmp) through the
SrrAB system, quickly and enzymatically hydrolyzes nitric oxide,
and resists the hosts inhibitory effect on pathogens (Richardson
et al., 2006); at the same time, Staphylococcus aureus upregulates
L-lactate dehydrogenase 1 (Ldh1), enabling it to survive lactic
acid fermentation under aerobic and anaerobic conditions
(Richardson et al., 2008). The virulence of Staphylococcus
aureus requires hexose produced by glycolysis, and an increase
in the glucose concentration will enhance the resistance of
pathogens to nitric oxide and subsequently the host immune
response (Vitko et al., 2015;Olive and Sassetti, 2016).
Polyamines are toxic to Staphylococcus aureus.Staphylococcus
aureus strains with arginine catabolic mobile element (ACME)
encode the acetyltransferase SpeG, which makes the strains
resistant to polyamines and facilitates colonization in host cells
(Diep et al., 2008;Olive and Sassetti, 2016). Pathogens evade
the killing effect of the hosts immune system, facilitating the
colonization of pathogens in the hosts reproductive tract. The
colonization of toxic shock syndrome toxin-1 (TSST-1)
Staphylooccus aureus strains will increase the production of
proinammatory cytokines and chemokines in human vaginal
epithelial cells, further destroying the mucosal barrier and
increasing the penetrating effect of TSST-1, leading to severe
symptoms and signs of vulvovaginitis (Pereira et al., 2013). This
also explains why vaginal inammation in AV is more serious
than that in BV. However, previous studies have mainly focused
on of BV-related bacteria, and there are few studies on AV-
related bacteria, metabolites, and immunity. Future research may
reveal the signicance if it is used as a future research direction.
NORMAL PREGNANCY
Unlike non-pregnant women, healthy pregnant women are
affected by estrogen-progesterone, and the vaginal microora
tends to be stable from the rst trimester to the third trimester,
that is, the low richness and low diversity dominated by
Lactobacillus spp. inhibits the growth of CST IV pathogenic
bacteria such as Gardnerella vaginalis, Atopobium vaginae,
Sneathia amnii,Prevotella Bivia,andPrevotella cluster 2
(MacIntyre et al., 2015;Brown et al., 2018;Serrano et al.,
2019). The vaginal ora during pregnancy is less transformed,
mostly between Lactobacillus species (Romero et al., 2014;
TABLE 3 | Current existing articles analyzing the correlation between the genital tract ora and metabolites in pregnant women.
Year Author Population Sample CST/
microorganisms
Metabolites
Normal pregnant women
2016 (Prince
et al.,
2016)
Women who delivered at term (n = 27), women
who delivered preterm (n = 44)
Placental
membranes
swabs
Bradyrhizobium
spp., streptococcus
thermophilus
Term cohorts: lower levels of the amino sugar and nucleotide
sugar metabolism, butanoate metabolism, riboavin
metabolism, and amino-benzoate degradation
2015 (McMillan
et al.,
2015)
Pregnant women (n = 67) and non-pregnant
women (n = 64)
Vaginal uid CST-I Similar with non-pregnant women :
Higher levels of succinate
CST-IV Similar with non-pregnant women :
1) Organic acid: higher levels of 2-hydroxyisovalerate,
g-hydroxybutyrate, 2-hydroxyglutarate and 2-
hydroxyisocaproate; lower levels of lactate
2) Higher levels of amines: tyramine, putrescine, and
cadaverine
3) Lower levels of amine precursors: tyrosine, lysine, ornithine
Preterm birth
2016 (Prince
et al.,
2016)
Women who delivered at term (n = 27), women
with spontaneous preterm birth (n = 44)
Placental
membranes
swabs
Lactobacillus
crispatus,
Acinetobacter
johnsonii
Preterm cohorts: higher levels of pentose phosphate pathway,
glycerophopholipid metabolism, and biosynthesis of the
siderophore group non-ribosomal peptides
2014 (Aagaard
et al.,
2014)
Women who delivered with preterm birth (n =
16), women with remote antenatal infection (n =
16), and controls (n = 16)
Placenta
tissue
Burkholderia spp. 1)Higher levels of methane metabolism, isoquinoline alkaloid
biosynthesis, and glycine/serine/threonine metabolism
2) Lower levels of biotin metabolism and
glycosylphosphatidylinositol anchor pathways
CST, Community state types.
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094888
Serrano et al., 2019). Pregnant women with CST I as the
dominant bacteria have the most stable vaginal ora
throughout pregnancy, followed by those with CST V, CST II,
and CST IV (MacIntyre et al., 2015). The vaginal microbiota
during the third trimester is similar to that of non-pregnant
women. One week after delivery, estrogen decreases, and
glycogen-supported Lactobacillus spp. also decreases sharply.
The stability and compliance of the vaginal ora decreased
signicantly, and the diversity increases, especially that of CST
IV, which leads to disorders of the postpartum vaginal ora and
even postpartum endometritis and puerperal morbidity
(DiGiulio et al., 2015;MacIntyre et al., 2015). There are also
microora in the placenta and amniotic uid of women who
experience normal-term delivery (Collado et al., 2016;Nuriel-
Ohayon et al., 2016). Some studies suggest that placental bacteria
may be derived from oral ora, mainly non-pathogenic
symbiotic ora such as Firmicutes,Tenericutes,Proteobacteria,
Bacteroidetes, and Fusobacteria phyla (Aagaard et al., 2014). The
origin of the placental microbiota is also related to the migration
of the intestinal ora to the fetus-placenta interface, which
promotes colonization of the fetus after birth (Collado et al.,
2016). However, some studies indicate that there are no
microorganisms in the normal placenta and amniotic uid,
which may be caused by the contamination of laboratory
reagents or equipment or by different methods used to
obtaining specimens (Leiby et al., 2018;Lim et al., 2018;De
Goffau et al., 2019).
Analysis of the metabolic characteristics of the vaginal
microbiota revealed that the microbial metabolic activity in the
rst trimester is the highest to adapt to changes in pregnancy
(Serrano et al., 2019)(Table 3). As pregnancy progresses, the
vaginal microbiota tends to become stable, and its metabolic
capacity tends to be simplied, mainly reected in the low
activity of carbohydrate metabolism, cell wall/membrane
biochemical pathways, protein synthesis pathways, and nucleic
acid metabolism pathways (Serrano et al., 2019). Another study
also found that the carbohydrate metabolism and lipid
metabolism of cervicovaginal secretions in full-term women
were downregulated during the second and third trimesters
(Ghartey et al., 2015). Carbohydrate metabolism was
signicantly downregulated, especially in the third trimester of
pregnancy, and was related to the large amount of glycogen
deposition and metabolizationintolacticacidinahighly
estrogen state (Ghartey et al., 2015). This change is conducive
to the colonization of lactobacilli in the host reproductive tract
and maintains the necessary acidic pH in the healthy
reproductive tract. It also helps maintain the integrity of the
cervix and is related to a reduction in adverse pregnancy
outcomes. In women who give birth at term, the lipid
metabolism of cervicovaginal secretions is signicantly reduced
in the third trimester, which may be related to the acidic
environment inhibiting the growth of pathogenic bacteria, and
the antimicrobial component of cervicovaginal secretions,
methyl-4-hydroxybenzoate, increases by approximately 8.8
times from the second to the third trimester, helping maintain
a stable vaginal microenvironment (Ghartey et al., 2015). The
metabolic pathways of amniotic uid and placental ora are
mainly involved in membrane transport, carbohydrate
metabolism, amino acid metabolism and energy metabolism,
which are closely related to the life activities of the fetal placenta
(Aagaard et al., 2014;Collado et al., 2016). Therefore, the
metabolic function of the genital tract ora during pregnancy
is of great signicance for maintaining pregnancy stability.
In normal pregnancy, the mother has increased immune
tolerance to fetal-expressed paternal antigens through
extended-selfantigens to maintain the growth of the fetus in
the body (Bromeld et al., 2017;Deshmukh and Way, 2019).
Maternal forkhead box P3 (FOXP3) Treg cells expand locally at
the maternal-fetal interface and expand systemically during
pregnancy to maintain allogeneic fetal tolerance (Agostinis et al.,
2019;Deshmukh and Way, 2019;Ghaemi et al., 2019). Metabolites
are closely related to host immunity during pregnancy. In humans,
the metabolism of L-arginine is related to the temporary
suppression of the maternal immune response during pregnancy
(Kropf et al., 2007). The activity of arginase expressed in the full-
term placenta of pregnant women increases signicantly, and the
high enzyme activity leads to a decrease in its substrate L-arginine,
which in turn induces the downregulation of T-cell receptor
(TCR) associated z-chain (CD3z) and the hyporesponsiveness of
functional T cells (Ismail, 2018). IDO also uses a similar approach
to silence T cells to induce and maintain immune tolerance (Kropf
et al., 2007). The normal reproductive tract ora plays an
important role in the establishment and consolidation of
mother-placental-fetal immunity to resist the interference of
external pathogenic bacteria (Mei et al., 2019). Studies have
demonstrated a correlation between Bacteroides species and
TCRgd+ T cells (participating in mucosal immunity) (Ghaemi
et al., 2019). The microbiota can induce the accumulation of Treg
cells, which are essential for maintaining immune tolerance, timely
endometrial receptivity, and correct placental implantation
(Benner et al., 2018). However, dysbiosis in the reproductive
genital tract can lead to immune disorders and participate in the
occurrence of adverse pregnancy outcomes (Smith and Ravel,
2017). Therefore, a comprehensive interpretation of the
reproductive tract microbes, metabolism, and immunity during
normal pregnancy provides a reference for discovering the causes
and mechanisms of adverse pregnancy outcomes (Wang
et al., 2016).
PREGNANCY-RELATED ADVERSE
OUTCOMES
Spontaneous Abortion and Infertility
Statistics from the Centers for Disease Control in the United
States showed that among married women aged 1544 years, 6%
hadinfertilityand12%hadimpairedfecundity,andthe
incidence increased yearly (Prevention, 2019). RTI is a risk
factor leading to reproductive dysfunction (such as infertility,
miscarriage, and repeated fertility failures), which in turn leads to
a clinical pregnancy rate of only 29.743.3% with embryo
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 6094889
transfer technology (Baker et al., 2010;Franasiak et al., 2016;
Koedooder et al., 2019). For example, the prevalence of BV in
infertile women is 1928%, while the clinical pregnancy success
rate is only 8% (Haahr et al., 2016;Bracewell-Milnes et al., 2018).
This observation may be related to abnormal vaginal microbiota
and pelvic pathogens (such as C. trachomatis) ascending to the
upper genital tract through the cervix, leading to PID and
reduced fertility (Witkin et al., 1995;Franasiak et al., 2016;
Haahr et al., 2016).
The normal reproductive tract ora provides a favorable
environment for embryo implantation, which can increase the
success rate and live birth rate of in vitro fertilization-embryo
transfer (IVFET) (Hyman et al., 2012;Sirota et al., 2014;
Koedooder et al., 2019). The genital tract microbiota in females
with fertility disorders is mainly manifested as a decrease in
Lactobacillus spp., an increase in non-Lactobacillus spp., a high
concentration of Candida spp., and an increase in the prevalence
of asymptomatic BV (Franasiak et al., 2016;Babu et al., 2017;
Campisciano et al., 2017;Wee et al., 2018;Koedooder et al.,
2019). The live birth rate will be reduced if harmful bacteria, such
as Gardnerella vaginalis,Atopobium vaginae,Acidovorax spp.,
Enterococcus spp., and Streptococcus spp., are found in the lower
reproductive tract (Hyman et al., 2012;Haahr et al., 2016;Wee
et al., 2018;Koedooder et al., 2019). Studies have shown that
before spontaneous abortion, endometrial aspiration uid has
higher bacterial diversity and lower Lactobacillus abundance
than before a healthy pregnancy (Moreno et al., 2020). The
presence of CST IV microbiota in the endometrium is related to
asignicant reduction in the incidence of implantation,
pregnancy, and continuous pregnancy (Moreno et al., 2016). In
2016, Verstraelen et al. (2016) performed endometrial biopsy on
19 women with fertility disorders (infertility, repeated
implantation failures, and repeated miscarriages) and found
that 90% of women with fertility disorders mainly had
Bacteroides phylum as the dominant bacteria in their
endometrium. In addition, when Gardnerella and Streptococcus
genra are detected in the endometrium, they have a particularly
adverse effect on reproductive outcomes (Moreno et al., 2016).
The endometrium microbiota may be carried by sperm and affect
the microbial composition of the female reproductive tract
(Koedooder et al., 2019). For example, when the detection rate
of Mycoplasma hominis, Neisseria genus, Klebsiella genus and
Pseudomonas genus in semen increases, it is not only related to a
low sperm concentration, abnormal sperm morphology, high
semen viscosity, and oligospermia but also indirectly leads to a
decline in female fertility (Ahmadi et al., 2017;Monteiro et al.,
2018). Microorganisms also colonize in the follicular uid, and
the low success rate of embryo transfer is related to the
colonization of Propionibacterium spp. and Streptococcus spp.
in the follicular uid (Pelzer et al., 2013). Therefore, the normal
genital tract ora is of great signicance to the maintenance
of fertility.
Endometrial receptivity and follicle quality in people with
reproductive disorders are closely related to metabolites in the
reproductive tract. Lipid homeostasis is essential for maintaining
health (Braga et al., 2019;Hernandez-Vargas et al., 2020). In
2019, Braga et al. (2019) analyzed the lipid metabolism of the
endometrial secretions taken from patients with IVFET cycles
before transplantation and found that phosphoethanolamine,
phosphatidic acid, diacylglycerol, triacylglycerol, glycosyl
diacylglycerol, phosphatidylcholine, neutral sphingolipid,
and lysophosphatidylglycerol are possible biomarkers of
endometrial receptivity and are associated with implantation
failure (Altmäe et al., 2014). Follicular uid is the
microenvironment for the growth of oocytes, and the
metabolism of follicular uid indirectly affects the growth
and development of oocytes (Bracewell-Milnes et al., 2017). In
2019, Song et al. (2019) conducted a targeted metabolomics
analysis of the follicular uid of patients with recurrent
spontaneous abortion after IVFET treatment and found that
eight metabolites, namely, dehydroepiandrosterone,
lysophosphatidylcholine (lysoPC) (16:0), lysoPC(18:2), lysoPC
(18:1), lysoPC(18:0), lysoPC(20:5), lysoPC(20:4), and lysoPC
(20:3) were upregulated in the recurrent abortion group, and
10 metabolites, namely, phenylalanine, linoleate, oleic acid,
docosahexaenoic acid, lithocholic acid, 25-hydroxyvitamin D3,
hydroxycholesterol, 13-hydroxy-alpha-tocopherol, leucine, and
tryptophan were downregulated. The above indicators can also
predict the success rate of transplantation. Therefore, it is very
meaningful to analyze metabolites in people with reproductive
disorders. However the metabonomic analysis of cervicovaginal
secretions in women with fertility disorders still needs more
research to fully prove the role of metabolism in fertility disorders
and the interaction between immunity and the ora.
Fertility dysfunction may be related to the destruction of
immune tolerance caused by a decline in the number and
function of Treg cells (Deshmukh and Way, 2019).Through
endometrial biopsies of women with infertility in the mid-
secretory phase of the menstrual cycle, it was found that the
expression of Foxp3 mRNA was reduced, suggesting that the
differentiation of uterine T cells into a Treg cell phenotype is
impaired, which may lead to reduced endometrial receptivity
(Jasper et al., 2006). Moreover, immunoglobulin-like transcript 4
+ (ILT4+) DCs may be involved in the process of recurrent
miscarriage and recurrent implantation failure induced by Foxp3
+ Treg cells (Liu et al., 2018). A reduction in maternal Treg cell
inhibitory ability caused by microbial infection can also cause
placental inammation, leading to the release and activation of
fetal-specic maternal CD8+ T cells, which inltrate the
decidua and lead to abortion (Deshmukh and Way, 2019). The
microbiota is important for basic CCL2 (monocyte chemotactic
protein-1, MCP-1) secretion to control the homeostasis of
plasmacytoid DCs, macrophage recruitment and polarization,
and local T cell balance (Sierra-Filardi et al., 2014;Swiecki et al.,
2017). DCs are a key regulator of immune tolerance during
pregnancy. Patients with elevated dehydroepiandrosterone and
dehydroepiandrosterone sulfate (DHEAS) in the follicular uid
have DC damage, which can cause infertility or spontaneous
abortion by causing the abnormal immunity of oocytes or
embryos (Song et al., 2019). When combined with a bacterial
ora disorder, it may aggravate the dysfunction of DCs, and
reproductive dysfunction is likely. It is known that plasma
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948810
tryptophan metabolism is closely related to abortion (Fei et al.,
2016). While BV-related bacteria are involved in the metabolism
of tryptophan and a variety of amino acids (Srinivasan et al.,
2015), it is necessary to further study whether genital tract
bacteria and their metabolites cause an imbalance of immune
tolerance, affect plasma metabolite levels and participate in the
occurrence of reproductive dysfunction. Future research should
focus on the local immune effects of genital tract ora and
metabolites on people with reproductive dysfunction as the
main research direction to explore the impact of the three
interactions on reproductive disorders.
Preterm Birth
Every year, 15 million babies are born premature worldwide,
accounting for approximately 11% of the live birth population
(Blencowe et al., 2012). Preterm birth caused by ascending
genitourinary tract infection accounts for 4050% of all
preterm births (Goldenberg et al., 2008). CST-IV vaginal
microora is closely related to premature delivery (DiGiulio
et al., 2015;Dunlop et al., 2015;Workowski and Bolan, 2015;
Brown et al., 2018;Han et al., 2019). Additionally, studies have
conrmed that pregnant women with BV have an increased risk
of premature birth (Callahan et al., 2017;Anahtar et al., 2018;
Chu et al., 2018;Fettweis et al., 2019;Serrano et al., 2019).
Preterm birth is the second leading cause of neonatal death (Liu
et al., 2016). Premature babies are prone to diabetes, chronic
inammation and cardiovascular disease in the long term
(Snyers et al., 2020). Therefore, the prevention of premature
birth is the top priority of medical work.
The Human Microbiome Project Multi-Omic Microbiome
Study showed that L. crispatus decreased and that BVAB1,
Sneathia amnii, TM7-H1 (BVAB-TM7), and partial Prevotella
species increased in the rst and second trimesters of women
who deliver prematurely, thus, these factors can be used as
markers for predicting preterm birth (Fettweis et al., 2019).
Studies have also shown that the colonization of vaginal
Streptococcus agalactiae and Klebsiella pneumonia in the
second trimester is signicantly associated with late
miscarriage and very premature delivery (before 28 weeks)
(Son et al., 2018;Koedooder et al., 2019). Changes in the
cervicovaginal ora of women who deliver prematurely greatly
alter the metabolome and are involved in premature cervical
remodeling (Table 3). Ghartey et al. (2015;2017) found that
women with symptoms of preterm birth and eventually
spontaneous preterm birth (sPTB) have signicant changes in
cervicovaginal metabolites. Lipid metabolism and carbohydrate
metabolism in the cervicovaginal secretions of women who
deliver prematurely are signicantly upregulated, and peptide
levels are signicantly reduced (Ghartey et al., 2015;Ghartey
et al., 2017). Upregulation of lipid and carbohydrate pathways is
associated with positive energy utilization and may be related to
early cervical remodeling and sPTB microbiota utilization
(Ghartey et al., 2015;Ghartey et al., 2017). A decrease in
dipeptides may reect the decreased level of proteolysis
in women who deliver prematurely and changes in the
activities of proteases and are associated with asymptomatic
sPTB (Ghartey et al., 2015). In addition, the level of N-
acetylneuraminate in the cervix of women who deliver
prematurely increased signicantly (by 4.9 times), which may
be related to the increased afnity of cells for infection and
participate in host immunity (Ghartey et al., 2015).
Embryo development and growth depend to a large extent on
placental function, and the placental microbiome may affect fetal
and pregnancy outcomes. Chorioamnionitis and intrauterine
infection are closely related to premature delivery (Chu et al.,
2018). However, the specic source of infection may be the
ascending infection of BV bacteria (Fettweis et al., 2019),
the ascending carrying of sperm (Svenstrup et al., 2003), the
colonization of endometrial bacteria (Cowling et al., 1992;Chu
et al., 2018), the retrograde infection of salpingitis, and the blood-
borne infection of oral bacteria (Chu et al., 2018). Studies have
shown that the bacteria in the uterus of women who deliver
prematurely are mainly derived from vaginal bacteria, such as
Burkholderia taxa, which is signicantly enriched in the placenta
(Goldenberg et al., 2000;Aagaard et al., 2014). The metabolic
enrichment of the lipopolysaccharide biosynthetic pathway of
the microbiota in the placenta may be related to the expansion
and reproduction of the microbiota (Aagaard et al., 2014). The
premature birth rate of women with bacteria detected in
amniotic uid is higher, and the metabolomics of the amniotic
uid of women who deliver prematurely are signicantly altered,
contributing to the initiation of preterm birth (Menon et al.,
2014;Collado et al., 2016). Studies have shown that there are
ora on the fetal membranes and that the composition of the
fetal membranes is closely related to the degree of inammation
of chorioamnionitis (Prince et al., 2016). Analysis of the
metabolic function of the fetal membrane microbiome revealed
that a reduction in the pentose phosphate pathway and
glycerophospholipid metabolism is related to chorioamnionitis,
and a reduction in glycerophosopholipid metabolism will lead to
an increase in the production of arachidonic acid, which is
related to inammation and prostanoid synthesis and is
involved in premature birth (Prince et al., 2016).
Inammation and antimicrobial peptide reactions involved in
certain vaginal microorganisms play a role in destroying and
invading cervical mucus plugs or amniotic membranes and
ultimately trigger proinammatory reactions, leading to
premature delivery (Goldenberg et al., 2000;Yarbrough et al.,
2015;Smith and Ravel, 2017;Strauss et al., 2018). It is known
that Gardnerella vaginalis ascends to infect the amniotic membrane
and irritate the cervix, leading to premature delivery. Gardnerella
vaginalis may activate the NACHT, LRR and PYD domains-
containing protein 3 (NLRP3) inammasomes through monocyte
NLRs; then, NLRP3 binds to and cleaves caspase-1, induces IL-1b,
IL-18, and TNF-asecretion, and ultimately leads to premature
delivery (Vick et al., 2014). Metabolites are involved in the
occurrence and development of preterm labor. When L. iners and
BV-related microorganisms are increased in the vagina, the ratio of
D-type/L-type lactic acid decreases, and matrix metalloproteinase
(MMP-8) increases (Witkin et al., 2013;MacIntyre et al., 2015). This
process eventually leads to premature cervix maturation and
ascending infection of the amniotic membrane and thus,
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948811
premature delivery (Yoon et al., 2001). Four proinammatory
cytokines, eotaxin, IL-1b,IL-6andMIP-1b, are increased
signicantly in the vagina of women who deliver prematurely
(Fettweis et al., 2019). BVAB1, Sneathia amnii,TM7-H1,
Prevotella timonensis,andPrevotella buccalis are closely related to
the levels of cytokines in vaginal secretions (Fettweis et al., 2019).
Both BVAB1 and TM7-H1 can produce pyruvate, acetate, L-lactate
and propionate. These SCFAs reduce antimicrobial activity and
promote the production of host proinammatory cytokines and are
also involved in the occurrence of preterm labor (Fettweis et al.,
2019). Therefore, there is a close correlation between the microbiota,
metabolites and host immune status in reproductive tracts of
women who deliver prematurely, and future researchers need to
study the relevant pathogenesis. However, the relationship of
microbial metabolites in the reproductive tract, preterm delivery
immunity and the onset of preterm delivery still needs
further exploration.
GYNECOLOGICAL ONCOLOGY
Dysbiosisisrelatedtotumorcarcinogenicity(Ilhan et al., 2019;Scott
et al., 2019). An imbalance in specic microorganisms can lead to
host epithelial barrier dysfunction, genome integration,
genotoxicity, inammatory activation, immune abnormalities and
metabolic abnormalities, creating a microenvironment that allows
tumor growth and further leading to the occurrence, development
and/or transfer of gynecological malignancies (Scott et al., 2019;
Laniewski et al., 2020). Among them, inammation is the central
feature of carcinogenesis and the main carcinogenic mechanism
related to cancer (Scott et al., 2019). Microbial virulence factors can
induce chronic inammation in host tissues, stimulate cell
proliferation, cause cell proliferation disorders, and combine with
the failure of cell apoptosis to ultimately lead to a malignant
phenotype (Scott et al., 2019;Laniewski et al., 2020). Metabolic
changes in cancer are the core of tumorigenesis and phenotypic
changes (Buckendahl et al., 2011;Zhang et al., 2012;Turkoglu et al.,
2016;Hopkins and Meier, 2017;Icard et al., 2018). Human
microorganisms are also involved in the formation of
carcinogenic metabolites and even exert genotoxicity to cause host
deoxyribonucleic acid (DNA) damage and participate in tumor
carcinogenicity (Kassie et al., 2001;Scott et al., 2019). Flora disorders
can also destroy the host-based anticancer immune monitoring to
promote tumor development and progression (Di Pietro et al., 2018;
Klein et al., 2019;Scott et al., 2019;Laniewski et al., 2020). Therefore,
the balance of the microenvironment of the reproductive tract has a
positive effect on maintaining the stability of the microbiota and
antitumor effects.
Cervical Intraepithelial Lesions and
Cervical Cancer
Cervical cancer (CC) is the most common human papillomavirus
(HPV)-related malignant tumor and the fourth most common
malignant tumor in women worldwide. In 2018, there were an
estimated 570,000 new cases and 311,000 deaths from this disease
(Bray et al., 2018). Approximately 85-90% of high-risk HPV
infections can be cleared spontaneously, and only 10-15% that
persist lead to cervical intraepithelial neoplasia (CIN) and invasive
cervical cancer (ICC). HPV-16 and HPV-18 are the main pathogens
of CC (Chase et al., 2015). The surface of the cervical mucosa is
susceptible to environmental inuences. When dysbiosis occurs, the
local cervicovaginal microenvironment may promote the
progression of malignant tumors together with HPV (Ma et al.,
2014;Laniewski et al., 2018;Chorna et al., 2020).
A decrease in Lactobacillus spp. and an increase in vaginal pH
are closely related to HPV infection, cervical lesions and CC
(Laniewski et al., 2018;Ilhan et al., 2019;Laniewski et al., 2020).
Cervical squamous intraepithelial lesions or CC also increase the
TABLE 4 | Current existing articles analyzing the correlation between the genital tract ora and metabolites in women with cervical intraepithelial lesions and cervical
cancer.
Year Author Population Sample CST Metabolites
2020 (Borgogna
et al.,
2020)
HPV-negative participants (n = 13) and HPV-positive
participants (n = 26)
Vaginal swabs CST-I Higher concentrations of histamine, 3-n-acetyl-LL-cysteine-
S-yl acetaminophen, and gammaaminobutyrate
CST-III High levels of 3-n-acetyl-L-cysteine-S-yl acetaminophen
CST-IV Low levels of heme, glycerophosphorylcholine, and
oxidized glutathione
2019 (Ilhan
et al.,
2019)
78 premenopausal, non-pregnant women and grouped as
follows: healthy HPV-negative (n = 18) and HPV-positive
participants (n = 11), low-grade squamous intraepithelial
lesions (n = 12), high-grade squamous intraepithelial lesions
(n = 27) and invasive cervical carcinoma (n = 10)
Cervicovaginal
lavages and
vaginal swabs
CST-IV 1) Higher levels of cadaverine, putrescine, tyramine,
tryptamine, agmatine, and glutathione synthesis
intermediate, 2-hydroxybutyrate, branched chain amino
acid metabolism product, alpha-hydroxy-isovalerate, and
L-isoleucine metabolism product, 2-hydroxy-3-methyl-
valerate
2) Lower levels of nucleotides adenosine and cytosine and
xenobiotics such as 2-keto-3-deoxy-gluconate and 1,2,3-
benzenetriol
2019 (Kwon
et al.,
2019)
Normal women (n = 18), cervical intraepithelial neoplasia two
or three patients (n = 17), and cervical cancer patients
(n = 12)
Cervical swabs CST-IV 1) Cervical cancer patients: enriched in peptidoglycan
biosynthesis (ko00550) pathway
2) Cervical intraeptithelia neoplasia 2/3 patients: enriched in
ko00300 (lysine biosynthesis), ko00680 (methane
metabolism), and ko05211 (renal cell carcinoma)
CST, Community state types; HPV, human papillomavirus.
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948812
diversity of the vaginal ora, limited not only to BV-related
microorganisms but also to non-BV bacteria, such as
Streptococcus agalactiae,Clostridium spp., Pseudomonadales order,
and Staphylococcus spp. (Klein et al., 2019;Laniewski et al., 2020).
An increase in the CIN stage is also related to an increase in the
diversity of the vaginal microbiota, suggesting that microorganisms
play a role in the regulation of persistent viral infection and disease
progression (Van Ostade et al., 2018). Compared with patients with
low-grade squamous intraepithelial lesions (LSILs), Sneathia
sanguinegens,Anaerococcus tetradius and Peptostreptococcus
anaerobius in patients with high-grade squamous intraepithelial
lesions (HSILs) are more enriched in the vagina, and
Mycoplasmatales order, Pseudomonadales order, and
Staphylococcus spp.aremoreenrichedinthecervix(Mitra et al.,
2015;Klein et al., 2019;Laniewski et al., 2020). Another study found
that Sneathia spp. and Fusobacterium spp. exist only in women with
cervical lesions or cancer but not in women without lesions
(Audirac-Chalifour et al., 2016). Therefore, the presence of
Sneathia in the vaginal microbiome may be a characteristic
microorganism of cervical lesions and CC (Laniewski et al., 2018;
Laniewski et al., 2020). Analysis of the metabolic pathways of the
bacterial ora in patients with CC showed that the peptidoglycan
biosynthesis (ko00550) pathway is signicantly enriched (Kwon
et al., 2019). Research has also found that the bacterial cell wall
peptidoglycan is not only essential for maintaining the overall
antiosmotic pressure of the bacteria to ensure cell survival but
also participates in the occurrence of inammation, affecting the
function of host neutrophils and the innate immune response.
Therefore, cervical microbes may promote the development of CC
and precancerous lesions by acting as a modulators of host
inammatory pathways.
The vaginal metabolism characteristics of HPV-infected and
uninfected patients are different, and the vaginal CST status drives
the metabolic characteristics of HPV-infected patients (Borgogna
et al., 2020)(Table 4). In vaginal CST III, HPV-infected women
have higher levels of biogenic amines than HPV uninfected women
(Borgogna et al., 2020). In CST IV, HPV-infected women have
lower concentrations of glutathione (GSH), oxidized glutathione
(GSSG), glycogen, and phospholipid-related metabolites than
uninfected women. There are also differences in the metabolic
characteristics of HPV infection, cervical lesions, and CC. Several
researchers conducted a study on the metabolome of cervicovaginal
secretions in HPV-mediated cervical tumors and found that
compared with HPV-negative group, HPV-positive group, and
cervical lesion group, the number and diversity of cervical vaginal
metabolites in CC patients were increased (Ilhan et al., 2019).
ComparedwiththeHPV-negativegroup,theHPV-positive,LSIL
and HSIL groups had fewer amino acids, and their metabolites in
cervical and vaginal secretions and the subpathways and depletion
levels under the amino acid superpathway were different (Ilhan
et al., 2019;Laniewski et al., 2020). In addition, in the vulvovaginal
secretions of patients with CC, volatile organic compounds, such as
alkanes, and methylated alkanes are different from those of healthy
women, which may be related to the oxidation of cell membrane
lipids and proteins during the carcinogenic process and the
production of volatile organic compounds (Rodriguez-Esquivel
et al., 2018;Ilhan et al., 2019). These metabolites can be used as
potential biomarkers for CC. In addition, the unique metabolic
characteristics in the cervicovaginal microenvironment can assist in
the diagnosis and differentiation of health, HPV infection
(Borgogna et al., 2020), HSILs, LSILs, and CC (Ilhan et al., 2019).
For example, long chain fatty acids, ketone bodies, steroids,
ceramides, and plasmalogens can distinguish individuals with ICC
from those with HPV ().
The interaction between the host and reproductive tract
microorganisms forms a metabolic network that participates in
the formation of the local tumor environment during the process
of continuous HPV infection and cancer progression (Ilhan et al.,
2019). In HSILs and CC, the vaginal microbial community
disrupts amino acid and nucleotide metabolism in a manner
similar to that in BV (Ilhan et al., 2019). Compared with healthy
individuals, the abundance of lipid metabolites in the vaginas of
ICC patients is higher (Ilhan et al., 2019;Szewczyk et al., 2019).
This phenomenon may be related to the interaction between
microbes and the host, which activates the carcinogenic
pathways in the tumor microenvironment and increases cell
proliferation and cell membrane synthesis, thus enhancing
the carcinogenic activity of the microora. Changes in the
cervicovaginal microbial community cannot only change the
cervicovaginal metabolome but also further affect immunity
and participate in cancer progression (Ilhan et al., 2019). For
example, glycochenodeoxycholate (GCDC) is a key metabolite of
host-Lactobacillus cometabolism and can inhibit vaginal ora
disorders (Ilhan et al., 2019). A decrease in GCDC and
Lactobacillus species in CC patients leads to a decrease in the
ability to induce inammation and toxic reactions and further
leads to a weakened antitumor effect.
The interaction between immunity and metabolites forms a
special tumor microenvironment. In the CIN group, the
concentrations of IL-8, IL-10, and nitric oxide in cervicovaginal
secretions were higher than those in the control group, indicating
that these mediators play a role in the tumor immune
microenvironment (Tavares-Murta et al., 2008). Since IL-8 is a
Th1-type cytokine and has a proinammatory effect and IL-10 is a
Th2-type cytokine and has an anti-inammatory effect (Fernandes
et al., 2015), the interaction mechanism between nitric oxide and the
two needs to be further studied. ICC patients with high genital
inammation (high IL-1a,IL-1b,IL-8,MIP-1b,CCL20,regulation
on activation normal T-cell expressed and secreted (RANTES), and
TNFaexpression) had the strongest correlation with lipids. An
increase in plasmalogens and long chain polyunsaturated fatty acids
in ICC not only indicates abnormal cell metabolism but also has a
proinammatory cytokine precursors effect, inducing abnormal
gene expression and disordered cytokine production (Ilhan et al.,
2019). Metabolites are also closely related to CC progression and
tumor cell growth. CC is characterized by an immunosuppressive
microenvironment and Th2-type cytokines (Bedoya et al., 2014). In
females with CC, Th2-type cytokines (IL-10 and IL-13) induce the
expression of arginase (ASE), which converts L-arginine into L-
ornithine and polyamines, and a reduction in L-arginine is related
to the downregulation of the immune response, further promoting
tumor progression (Bedoya et al., 2014). Therefore, an increase in
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948813
polyamines in the vagina ora of CST IV HPV-positive patients is a
metabolic feature that HPV uses to escape host immunity and
promote tumor progression (Borgogna et al., 2020). More research
is needed to support the impact of HPV infection, cervical lesions,
and the direct mechanism of action between the bacterial ora,
metabolites and immunity in the cervicovaginal secretions of
patients with CC on tumor progression and tumor metastasis.
Endometrial Cancer
In 2018, there were an estimated 382,069 new cases and 89,929
deaths related to corpus uteri cancer (Bray et al., 2018).
Endometrial cancer (EC) is a perimenopausal and
postmenopausal tumor, divided into two categories: type I and
type II (Troisi et al., 2018). Type I EC is the most common (Troisi
et al., 2018). Environmental factors, including obesity,
inammation, postmenopausal estrogen metabolism imbalance
and estrogen therapy, are themainriskfactorsforthe
development of type I EC (Laniewski et al., 2020). Type II EC is
rare and is mainly related to endometrial atrophy (Troisi et al.,
2018). Environmental factors are related to changes in the
intestinal and vaginal microbiomes. The close relationship
between the ora, estrogen metabolism and obesity indicates the
potential role of the microbiome in the etiology of EC (Laniewski
et al., 2020). EC is also closely related to PID, and an imbalance in
the vaginal ora can cause PID through ascending infection, so an
imbalance in the vaginal ora may be indirectly related to EC
(Ness et al., 2005;Yang et al., 2015;Champer et al., 2018).
The reproductive tract microbiota is involved in the
pathogenesis of EC (Laniewski et al., 2020). In 2016, Walther-
Antonio et al. (2016) analyzed the genital tract ora of 17
patients with EC, four with endometrial hyperplasia and 10
with benign uterine diseases and found that in the EC cohort,
Porphyromonas sp. was common in the vagina and cervix,
Bacteroides and Faecalibacterium sp.werecommoninthe
endometrium, and Bacteroides sp.was common in the ovary.
The endometrial microbiota of the EC and hyperplasia cohorts
was similar but differed to some degree from that of the benign
cohort. The endometrial hyperplasia and benign cohorts had
different microbiota structures, indicating that the microbiota
plays a role in the early stages of cell transformation (Walther-
Antonio et al., 2016). EC patients usually have a high vaginal pH.
The detection of vaginal Atopobium vaginae and Porphyromonas
sp. combined with a high vaginal pH is statistically correlated
with EC (Walther-Antonio et al., 2016). Studies have found that
Atopobium vaginae can induce proinammatory cytokines and
antimicrobial peptides, cause chronic inammation and local
immune disorders, promote Porphyromonas sp. infection in
cells, destroy normal cell regulatory functions, and ultimately
lead to carcinogenic processes (Walther-Antonio et al., 2016;
Laniewski et al., 2020). The link between Atopobium vaginae and
Porphyromonas sp. supports the link between BV-related
bacteria, immunity and EC (Laniewski et al., 2020).
EC has a unique endometrial metabolic signature. In 2017,
Altadill et al. (2017) studied the metabolomics of endometrial
tissue samples from 39 EC patients and 17 healthy women and
found lipids, kynurenine, endocannabinoids and RNA editing
pathway disorders in EC patients. Through further research on
RNA editing pathways, we found that adenosine deaminases
acting on RNA2 (ADAR2) are overexpressed in EC and are
positively correlated with tumor aggressiveness. ADAR2 may
contribute to the carcinogenicity of EC and can be used as a
potential marker for EC treatment. However, whether the genital
tract ora metabolites involved in EC carcinogenesis remains to
be studied. It is known that the concentration of hydroxybutyric
acid in the reproductive tract of BV patients is elevated. In the
intestine, SCFAs (such as hydroxybutyrate) induce Treg cells
through histone deacetylases (HDACs) and exert an
immunosuppressive effect in innate immune cells (McMillan
et al., 2015;Chen and Stappenbeck, 2019). Whether
hydroxybutyrate in the reproductive tract induces immune
suppression through HDACs and promotes the growth of
endometrial tumors requires further in vitro experiments.
Ovarian Cancer
Ovarian cancer (OC) is one of the deadliest malignant tumors in
women and the main cause of death from gynecological
malignancies (Turkoglu et al., 2016;Laniewski et al., 2020). In
2018, an estimated 295,414 new cases of OC were diagnosed
worldwide, and 184,799 women died from the disease, ranking
fth among cancer-related deaths (Bray et al., 2018). More than 80%
of patients have advanced disease, and the ve-year overall survival
rate is between 15 and 45% (Turkoglu et al., 2016). Similar to EC,
chronic infection of sexually transmitted pathogens and ascending
infection of genital tract inammation are related to the occurrence
of OC (Shanmughapriya et al., 2012;Idahl et al., 2020).
In 2019, Nene et al. (2019) rst published a study on the
presence of abnormal uterine ora in women with OC or at risk
of OC and found a strong correlation of OC or the breast cancer
susceptibility gene 1 (BRCA1) mutation status with participants
aged <50 years and those with a non-Lactobacillus dominant
microbiota. Women who have used oral contraceptive pills or
combined hormones for more than 5 years are more likely to
have Lactobacillus dominance and a lower risk of OC than
women who are using oral contraceptive pills or women who
have used combined hormones for less than 5 years. Compared
with the healthy surrounding ovarian tissue of the same
individual, OC tissue has unique microbial characteristics
(Banerjee et al., 2017). Potentially pathogenic intracellular
microorganisms, such as Brucella spp., Chlamydia spp. and
Mycoplasma spp., are present in 6076% of ovarian tumors
(Banerjee et al., 2017). In addition, an increase in
Proteobacteria and Firmicutes phyla in ovarian tumors,
especially an increase in Actinobacteria phyla, may cause
double-stranded breaks by releasing bacterial toxins (such as
colibactin and cytolethal distending toxin) and directly damage
cellular DNA (Banerjee et al., 2017;Nene et al., 2019). In
addition, several pathogenic viruses, intracellular bacteria,
fungi and parasites also exist in ovarian tissue (Banerjee et al.,
2017). These microorganisms may induce cancer through direct
or indirect mechanisms (Laniewski et al., 2020). It is known that
the integration of the HPV genome into the human genome is an
important reason for the development of CC. In 2017,
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948814
Banerjee et al. (2017) found HPV signals in the tumor tissues of
OC patients, and there was also an integration phenomenon. For
example, HPV16 has the largest number of viral integration sites
in human chromosomes. It can be integrated into various
intronic regions and genetic regions within 56 kb upstream of
many cancer-related human genes. In addition, the coding
sequence of the E1 gene of HPV18 is integrated in the intronic
region of the non-coding RNA gene of the host chromosomes.
All of the above factors may lead to the dysregulation of gene
expression and participate in the occurrence and development of
cancer (Banerjee et al., 2017). This research provides new ideas
for exploring the molecular mechanism of OC.
It is known that the metabolic characteristics of OC cells, OC
tissues, and ascites are signicantly changed and are closely
related to tumor tissue energy utilization, reproductive tract
inammation, the invasion and migration of OC cells, and the
chemotherapy resistance of OC (Fong et al., 2011;Poisson et al.,
2015). However, there are still few studies on the correlation
between the characteristics of microbial metabolism in the
reproductive tract and OC. The ovary is the end organ of the
upper genital tract and is affected by the ascending bacteria of
the genital tract, the bacteria of the ovary and the ora in the
abdominal cavity. Whether the metabolic characteristics of the
above bacteria are related to the occurrence and development of
ovarian tumors and the carcinogenicity of tumors, leading to a
unique tumor microenvironment, needs further research.
Metabolites are closely related to tumor immunity and tumor
development (Turkoglu et al., 2016;Clifford et al., 2018;
Szewczyk et al., 2019). Whether the metabolites of the genital
tract ora of OC patients participate in tumor immunity and
host antitumor immunity, which affects the occurrence,
development and metastasis of OC, still needs further research.
CONCLUSION
The host reproductive tract microenvironment includes
microorganisms, metabolites and immunity, and the balance of
the interactions among them is essential to maintain reproductive
health. Existing research on the relationship between female
reproductive tract microbes or immunity and reproductive tract
inammation, pregnancy, and tumors is becoming increasingly
detailed. In contrast to research on intestinal metabolites, research
on female reproductive tract metabolites is still in the preliminary
stage. Moreover, the reproductive tract metabolic characteristics of
AV, reproductive dysfunction, EC, and OC still need more research
at present, as relevant data are lacking.Additionally,therelationship
between microbial metabolites and host immunity in inammation,
pregnancy, and tumors of the female reproductive tract is relatively
unclear, and it may become a new direction for future research.
According to the current research, BV-related/CST IV bacteria and
the microenvironment formed by the reproductive tract have the
most comprehensive research on the adverse pregnancy outcomes
and tumor pathogenesis. The reproductive tract microenvironment
produced by BV/CST IV not only participates in the ascending
infection causing PID, which leads to an increased risk of
infertility, miscarriage, and premature delivery, but also
participates in the increased risks of precancerous lesions and
malignancy of the reproductive tract. The pathogenic mechanism
of adverse pregnancy outcomes and tumor diseases in the AV
microenvironment of the reproductive tract needs to be further
explored. Molecular detection technology combined with immune
and metabolomics can be used to better describe the function and
metabolic status of the ora, infer the possible pathogenic pathways
and immune response status, and analyze the complicated
relationship of the local microenvironment with inammation,
pregnancy, and tumor diseases. This method is also the best way
to study the pathogenic mechanism and disease characteristics of
female reproductive tract inammation, pregnancy, and
tumor diseases in the reproductive tract microenvironment. The
study of microorganisms, metabolites, and immunity in the
microenvironment of the reproductive tract under different
diseases (and then the development of targeted therapies for the
above three) was the main purpose of this article. At present, there
are many studies on the treatment of microorganisms with
antibiotics and probiotics. Studies have proven that probiotic
supplementation is very helpful in reducing the risk of
inammation, adverse pregnancy outcomes, and cancers (Nene
et al., 2019;Laniewski et al., 2020) and improving the ability to
respond to cancer treatments and quality of life (Postler and Ghosh,
2017;Champer et al., 2018;Laniewski et al., 2020). However,
microbial therapy is still prone to relapse and associated with a
high risk of recurrence. Whether targeted therapy for metabolites
(Sevin et al., 2015) and immunity (Ventriglia et al., 2017;Deshmukh
and Way, 2019) can be used to treat diseases or improve the effect of
microbial therapy still needs further research. Therefore, studying
theroleandmechanismofreproductivetractora, metabolites, and
immunity in disease pathogenesis will aid in disease diagnosis and
treatment and improve female reproductive health.
AUTHOR CONTRIBUTIONS
HL, CH, and FX conceived the study question, and all authors
were involved in the study design. HL created the rst draft of the
manuscript.YZ,CW,HYL,andAFmadesubstantial
contributions to drafting the article and revising it
critically. All authors contributed to the article and approved
the submitted version.
FUNDING
This work was supported by Tianjin Municipal Science and
Technology Commission Special Foundation for Science and
Technology Major Projects in Control and Prevention of Major
Diseases (Grant No. 18ZXDBSY00200), General Project of the
National Natural Science Foundation of China (Grant No.
82071674) and Tianjin Health Science and Technology Project
(Grant No. KJ20003).
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948815
REFERENCES
Aagaard, K., Ma, J., Antony, K. M., Ganu, R., Petrosino, J., and Versalovic, J.
(2014). The placenta harbors a unique microbiome. Sci. Transl. Med. 6,
237ra265. doi: 10.1126/scitranslmed.3008599
Abu-Lubad, M., Meyer, T. F., and Al-Zeer, M. A. (2014). Chlamydia trachomatis
inhibits inducible NO synthase in human mesenchymal stem cells by
stimulating polyamine synthesis. J. Immunol. 193, 29412951. doi: 10.4049/
jimmunol.1400377
Agostinis, C., Mangogna, A., Bossi, F., Ricci, G., Kishore, U., and Bulla, R. (2019).
Uterine Immunity and Microbiota: A Shifting Paradigm. Front. Immunol. 10,
2387. doi: 10.3389/mmu.2019.02387
Agrawal, T., Bhengraj, A. R., Vats, V., Salhan, S., and Mittal, A. (2011). Expression
of TLR 2, TLR 4 and iNOS in cervical monocytes of Chlamydia trachomatis-
infected women and their role in host immune response. Am. J. Reprod.
Immunol. 66, 534543. doi: 10.1111/j.1600-0897.2011.01064.x
Ahmadi, M. H., Mirsalehian, A., Sadighi Gilani, M. A., Bahador, A., and Talebi, M.
(2017). Asymptomatic Infection With Mycoplasma hominis Negatively Affects
Semen Parameters and Leads to Male Infertility as Conrmed by Improved
Semen Parameters After Antibiotic Treatment. Urology 100, 97102. doi:
10.1016/j.urology.2016.11.018
Aiyar, A., Quayle, A. J., Buckner, L. R., Sherchand, S. P., Chang, T. L., Zea, A. H.,
et al. (2014). Inuence of the tryptophan-indole-IFNgaxis on human genital
Chlamydia trachomatis infection: role of vaginal co-infections. Front. Cell
Infect. Microbiol. 4, 72. doi: 10.3389/fcimb.2014.00072
Aldunate, M., Srbinovski, D., Hearps, A. C., Latham, C. F., Ramsland, P. A.,
Gugasyan, R., et al. (2015). Antimicrobial and immune modulatory effects of
lactic acid and short chain fatty acids produced by vaginal microbiota
associated with eubiosis and bacterial vaginosis. Front. Physiol. 6, 164. doi:
10.3389/fphys.2015.00164
Al-Mushrif, S., Eley, A., and Jones, B. M. (2000). Inhibition of chemotaxis by organic
acids from anaerobes may prevent a purulent response in bacterial vaginosis. J. Med.
Microbiol. 49, 10231030. doi: 10.1099/0022-1317-49-11-1023
Altadill, T., Dowdy, T. M., Gill, K., Reques, A., Menon, S. S., Moiola, C. P., et al.
(2017). Metabolomic and Lipidomic Proling Identies The Role of the RNA
Editing Pathway in Endometrial Carcinogenesis. Sci. Rep. 7, 8803. doi: 10.1038/
s41598-017-09169-2
Altmäe, S., Esteban, F. J., Stavreus-Evers, A., Simon, C., Giudice, L., Lessey, B. A.,
et al. (2014). Guidelines for the design, analysis and interpretation of omics
data: focus on human endometrium. Hum. Reprod. Update 20, 1228. doi:
10.1093/humupd/dmt048
Anahtar, M. N., Gootenberg, D. B., Mitchell, C. M., and Kwon, D. S. (2018).
Cervicovaginal Microbiota and Reproductive Health: The Virtue of Simplicity.
Cell Host Microbe 23, 159168. doi: 10.1016/j.chom.2018.01.013
Audirac-Chalifour, A., Torres-Poveda, K., Bahena-Roman, M., Tellez-Sosa, J.,
Martı
nez-Barnetche, J., Cortina-Ceballos, B., et al. (2016). Cervical
Microbiome and Cytokine Prole at Various Stages of Cervical Cancer: A
Pilot Study. PloS One 11, e0153274. doi: 10.1371/journal.pone.0153274
Babu, G., Singaravelu, B. G., Srikumar, R., Reddy, S. V., and Kokan, A. (2017).
Comparative Study on the Vaginal Flora and Incidence of Asymptomatic
Vaginosis among Healthy Women and in Women with Infertility Problems of
Reproductive Age. J. Clin. Diagn. Res. 11, Dc18dc22. doi: 10.7860/JCDR/
2017/28296.10417
Baker, V. L., Jones, C. E., Cometti, B., Hoehler, F., Salle, B., Urbancsek, J., et al.
(2010). Factors affecting success rates in two concurrent clinical IVF trials: an
examination of potential explanations for the difference in pregnancy rates
between the United States and Europe. Fertil. Steril. 94, 12871291. doi:
10.1016/j.fertnstert.2009.07.1673
Banerjee, S., Tian, T., Wei, Z., Shih, N., Feldman, M. D., Alwine, J. C., et al. (2017).
The ovarian cancer oncobiome. Oncotarget 8, 3622536245. doi: 10.18632/
oncotarget.16717
Baqui, A. H., Lee, A. C. C., Kof, A. K., Khanam, R., Mitra, D. K., Dasgupta, S. K.,
et al. (2019). Prevalence of and risk factors for abnormal vaginal ora and its
association with adverse pregnancy outcomes in a rural district in north-east
Bangladesh. Acta Obstet. Gynecol. Scand. 98, 309319. doi: 10.1111/aogs.13492
Beatty, W. L., Belanger, T. A., Desai, A. A., Morrison, R. P., and Byrne, G. I. (1994).
Tryptophan depletion as a mechanism of gamma interferon-mediated
chlamydial persistence. Infect. Immun. 62, 37053711. doi: 10.1128/
IAI.62.9.3705-3711.1994
Bedoya, A. M., Tate, D. J., Baena, A., Cordoba, C. M., Borrero, M., Pareja, R., et al.
(2014). Immunosuppression in cervical cancer with special reference to
arginase activity. Gynecol. Oncol. 135, 7480. doi: 10.1016/j.ygyno.2014.07.096
Belland, R. J., Nelson, D. E., Virok, D., Crane, D. D., Hogan, D., Sturdevant, D.,
et al. (2003). Transcriptome analysis of chlamydial growth during IFN-
gamma-mediated persistence and reactivation. Proc. Natl. Acad. Sci. U.S.A.
100, 1597115976. doi: 10.1073/pnas.2535394100
Benner, M., Ferwerda, G., Joosten, I., and Van Der Molen, R. G. (2018). How
uterine microbiota might be responsible for a receptive, fertile endometrium.
Hum. Reprod. Update 24, 393415. doi: 10.1093/humupd/dmy012
Blencowe, H., Cousens, S., Oestergaard, M. Z., Chou, D., Moller, A. B., Narwal, R.,
et al. (2012). National, regional, and worldwide estimates of preterm birth rates
in the year 2010 with time trends since 1990 for selected countries: a systematic
analysis and implications. Lancet 379, 21622172. doi: 10.1016/S0140-6736
(12)60820-4
Borgogna, J. C., Shardell, M. D., Santori, E. K., Nelson, T. M., Rath, J. M., Glover, E.
D., et al. (2020). The vaginal metabolome and microbiota of cervical HPV-
positive and HPV-negative women: a cross-sectional analysis. Bjog 127, 182
192. doi: 10.1111/1471-0528.15981
Bracewell-Milnes, T., Saso, S., Abdalla, H., Nikolau, D., Norman-Taylor, J.,
Johnson, M., et al. (2017). Metabolomics as a tool to identify biomarkers to
predict and improve outcomes in reproductive medicine: a systematic review.
Hum. Reprod. Update 23, 723736. doi: 10.1093/humupd/dmx023
Bracewell-Milnes, T., Saso, S., Nikolaou, D., Norman-Taylor, J., Johnson, M., and
Thum, M. Y. (2018). Investigating the effect of an abnormal cervico-vaginal
and endometrial microbiome on assisted reproductive technologies: A
systematic review. Am. J. Reprod. Immunol. 80, e13037. doi: 10.1111/aji.13037
Braga, D., Borges, E.Jr., Godoy, A. T., Montani, D. A., Setti, A. S., Zanetti, B. F.,
et al. (2019). Lipidomic prole as a noninvasive tool to predict endometrial
receptivity. Mol. Reprod. Dev. 86, 145155. doi: 10.1002/mrd.23088
Bray, F., Ferlay, J., Soerjomataram, I., Siegel, R. L., Torre, L. A., and Jemal, A.
(2018). Global cancer statistics 2018: GLOBOCAN estimates of incidence and
mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68,
394424. doi: 10.3322/caac.21492
Bromeld, J. J., Rizo, J. A., and Ibrahim, L. A. (2017). Paternal priming of maternal
tissues to optimise pregnancy success. Reprod. Fertil. Dev. 30, 5055. doi:
10.1071/RD17345
Brown, R. G., Marchesi, J. R., Lee, Y. S., Smith, A., Lehne, B., Kindinger, L. M., et al.
(2018). Vaginal dysbiosis increases risk of preterm fetal membrane rupture,
neonatal sepsis and is exacerbated by erythromycin. BMC Med. 16, 9. doi:
10.1186/s12916-017-0999-x
Brunham, R. C., and Rey-Ladino, J. (2005). Immunology of Chlamydia infection:
implications for a Chlamydia trachomatis vaccine. Nat. Rev. Immunol. 5, 149
161. doi: 10.1038/nri1551
Buckendahl, A. C., Budczies, J., Fiehn, O., Darb-Esfahani, S., Kind, T., Noske, A.,
et al. (2011). Prognostic impact of AMP-activated protein kinase expression in
ovarian carcinoma: correlation of protein expression and GC/TOF-MS-based
metabolomics. Oncol. Rep. 25, 10051012. doi: 10.3892/or.2011.1162
Budilovskaya, O. V., Shipitsina, E. V., Spasibova, E. V., Pereverzeva, N. A.,
Vorobeva, N. E., Tsypurdeeva, N. D., et al. (2020). Differential Expression of
Local Immune Response Genes in the Vagina: Implication for the Diagnosis of
Vaginal Infections. Bull. Exp. Biol. Med. 168, 646650. doi: 10.1007/s10517-
020-04771-3
Byrne, G. I., Carlin, J. M., Merkert, T. P., and Arter, D. L. (1989). Long-term effects
of gamma interferon on chlamydia-infected host cells: microbicidal activity
follows microbistasis. Infect. Immun. 57, 13181320. doi: 10.1128/
IAI.57.4.1318-1320.1989
Callahan, B. J., Digiulio, D. B., Goltsman, D. S. A., Sun, C. L., Costello, E. K.,
Jeganathan, P., et al. (2017). Replication and renement of a vaginal microbial
signature of preterm birth in two racially distinct cohorts of US women. Proc.
Natl. Acad. Sci. U.S.A. 114, 99669971. doi: 10.1073/pnas.1705899114
Campisciano, G., Florian, F., Deustacchio, A., Stankovic, D., Ricci, G., De Seta, F.,
et al. (2017). Subclinical alteration of the cervical-vaginal microbiome in
women with idiopathic infertility. J. Cell Physiol. 232, 16811688. doi:
10.1002/jcp.25806
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948816
Ceccarani, C., Foschi, C., Parolin, C., Dantuono, A., Gaspari, V., Consolandi, C.,
et al. (2019). Diversity of vaginal microbiome and metabolome during genital
infections. Sci. Rep. 9, 14095. doi: 10.1038/s41598-019-50410-x
Champer, M., Wong, A. M., Champer, J., Brito, I. L., Messer, P. W., Hou, J. Y.,
et al. (2018). The role of the vaginal microbiome in gynaecological cancer. Bjog
125, 309315. doi: 10.1111/1471-0528.14631
Chase, D., Goulder, A., Zenhausern, F., Monk, B., and Herbst-Kralovetz, M.
(2015). The vaginal and gastrointestinal microbiomes in gynecologic cancers: a
review of applications in etiology, symptoms and treatment. Gynecol. Oncol.
138, 190200. doi: 10.1016/j.ygyno.2015.04.036
Chen,C.,Song,X.,Wei,W.,Zhong,H.,Dai,J.,Lan,Z.,etal.(2017).Themicrobiota
continuum along the female reproductive tract and its relation to uterine-related
diseases. Nat. Commun. 8, 875. doi: 10.1038/s41467-017-00901-0
Chen, F., and Stappenbeck, T. S. (2019). Microbiome control of innate reactivity.
Curr. Opin. Immunol. 56, 107113. doi: 10.1016/j.coi.2018.12.003
Chorna, N., Romaguera, J., and Godoy-Vitorino, F. (2020). Cervicovaginal
Microbiome and Urine Metabolome Paired Analysis Reveals Niche
Partitioning of the Microbiota in Patients with Human Papilloma Virus
Infections. Metabolites 10, 36. doi: 10.3390/metabo10010036
Chu, D. M., Seferovic, M., Pace, R. M., and Aagaard, K. M. (2018). The
microbiome in preterm birth. Best Pract. Res. Clin. Obstet. Gynaecol. 52,
103113. doi: 10.1016/j.bpobgyn.2018.03.006
Clifford, C., Vitkin, N., Nersesian, S., Reid-Schachter, G., Francis, J. A., and Koti,
M. (2018). Multi-omics in high-grade serous ovarian cancer: Biomarkers from
genome to the immunome. Am. J. Reprod. Immunol. 80, e12975. doi: 10.1111/
aji.12975
Collado, M. C., Rautava, S., Aakko, J., Isolauri, E., and Salminen, S. (2016). Human
gut colonisation may be initiated in utero by distinct microbial communities in
the placenta and amniotic uid. Sci. Rep. 6, 23129. doi: 10.1038/srep23129
Consortium, H. M. P. (2012). Structure, function and diversity of the healthy
human microbiome. Nature 486, 207214. doi: 10.1038/nature11234
Cowling, P., Mccoy, D. R., Marshall, R. J., Padeld, C. J., and Reeves, D. S. (1992).
Bacterial colonization of the non-pregnant uterus: a study of pre-menopausal
abdominal hysterectomy specimens. Eur. J. Clin. Microbiol. Infect. Dis. 11,
204205. doi: 10.1007/BF01967084
De Goffau, M. C., Lager, S., Sovio, U., Gaccioli, F., Cook, E., Peacock, S. J., et al.
(2019). Human placenta has no microbiome but can contain potential
pathogens. Nature 572, 329334. doi: 10.1038/s41586-019-1451-5
Delgado-Diaz, D. J., Tyssen, D., Hayward, J. A., Gugasyan, R., Hearps, A. C., and
Tachedjian, G. (2019). Distinct Immune Responses Elicited From
Cervicovaginal Epithelial Cells by Lactic Acid and Short Chain Fatty Acids
Associated With Optimal and Non-optimal Vaginal Microbiota. Front. Cell
Infect. Microbiol. 9, 446. doi: 10.3389/fcimb.2019.00446
Deshmukh, H., and Way, S. S. (2019). Immunological Basis for Recurrent Fetal
Loss and Pregnancy Complications. Annu. Rev. Pathol. 14, 185210. doi:
10.1146/annurev-pathmechdis-012418-012743
Di Pietro, M., Filardo, S., Porpora, M. G., Recine, N., Latino, M. A., and Sessa, R.
(2018). HPV/Chlamydia trachomatis co-infection: metagenomic analysis of
cervical microbiota in asymptomatic women. New Microbiol. 41, 3441.
Diep, B. A., Stone, G. G., Basuino, L., Graber, C. J., Miller, A., Des Etages, S. A.,
et al. (2008). The arginine catabolic mobile el ement and staphylococcal
chromosomal cassette mec linkage: convergence of virulence and resistance
in the USA300 clone of methicillin-resistant Staphylococcus aureus. J. Infect.
Dis. 197, 15231530. doi: 10.1086/587907
DiGiulio, D. B., Callahan, B. J., Mcmurdie, P. J., Costello, E. K., Lyell, D. J.,
Robaczewska, A., et al. (2015). Temporal and spatial variation of the human
microbiota during pregnancy. Proc. Natl. Acad. Sci. U.S.A. 112, 1106011065.
doi: 10.1073/pnas.1502875112
Donders, G. G. G., Bellen, G., Grinceviciene, S., Ruban, K., and Vieira-Baptista, P.
(2017). Aerobic vaginitis: no longer a stranger. Res. Microbiol. 168, 845858.
doi: 10.1016/j.resmic.2017.04.004
Dunlop, A. L., Mulle, J. G., Ferranti, E. P., Edwards, S., Dunn, A. B., and Corwin,
E. J. (2015). Maternal Microbiome and Pregnancy Outcomes That Impact
Infant Health: A Review. Adv. Neonatal Care 15, 377385. doi: 10.1097/
ANC.0000000000000218
Fehlner-Gardiner, C., Roshick, C., Carlson, J. H., Hughes, S., Belland, R. J.,
Caldwell, H. D., et al. (2002). Molecular basis dening human Chlamydia
trachomatis tissue tropism. A possible role for tryptophan synthase. J. Biol.
Chem. 277, 2689326903. doi: 10.1074/jbc.M203937200
Fei, H., Hou, J., Wu, Z., Zhang, L., Zhao, H., Dong, X., et al. (2016). Plasma
metabolomic prole and potential biomarkers for missed abortion. BioMed.
Chromatogr. 30, 19421952. doi: 10.1002/bmc.3770
Fernandes, J. V., Ta, D. E. M. F., Jc, D. E. A., Cobucci, R. N., Mg, D. E. C., Andrade,
V. S., et al. (2015). Link between chronic inammation and human
papillomavirus-induced carcinogenesis (Review). Oncol. Lett. 9, 10151026.
doi: 10.3892/ol.2015.2884
Fettweis, J. M., Serrano, M. G., Brooks, J. P., Edwards, D. J., Girerd, P. H., Parikh,
H. I., et al. (2019). The vaginal microbiome and preterm birth. Nat. Med. 25,
10121021. doi: 10.1038/s41591-019-0450-2
Fong, M. Y., Mcdunn, J., and Kakar, S. S. (2011). Identication of metabolites in
the normal ovary and their transformation in primary and metastatic ovarian
cancer. PloS One 6, e19963. doi: 10.1371/journal.pone.0019963
Franasiak, J. M., Werner, M. D., Juneau, C. R., Tao, X., Landis, J., Zhan, Y., et al.
(2016). Endometrial microbiome at the time of embryo transfer: next-
generation sequencing of the 16S ribosomal subunit. J. Assist. Reprod. Genet.
33, 129136. doi: 10.1007/s10815-015-0614-z
France, M. T., Mendes-Soares, H., and Forney, L. J. (2016). Genomic Comparisons
of Lactobacillus crispatus and Lactobacillus iners Reveal Potential Ecological
Drivers of Community Composition in the Vagina. Appl. Environ. Microbiol.
82, 70637073. doi: 10.1128/AEM.02385-16
Gajer, P., Brotman, R. M., Bai, G., Sakamoto, J., Schutte, U. M., Zhong, X., et al.
(2012). Temporal dynamics of the human vaginal microbiota. Sci. Transl. Med.
4, 132ra152. doi: 10.1126/scitranslmed.3003605
Ghaemi, M. S., Digiulio, D. B., Contrepois, K., Callahan, B., Ngo, T. T. M., Lee-
Mcmullen, B., et al. (2019). Multiomics modeling of the immunome,
transcriptome, microbiome, proteome and metabolome adaptations during
human pregnancy. Bioinformatics 35, 95103. doi: 10.1093/bioinformatics/
bty537
Ghartey, J., Bastek, J. A., Brown, A. G., Anglim, L., and Elovitz, M. A. (2015).
Women with preterm birth have a distinct cervicovaginal metabolome. Am. J.
Obstet. Gynecol. 212, 776.e771776.e712. doi: 10.1016/j.ajog.2015.03.052
Ghartey, J., Anglim, L., Romero, J., Brown, A., and Elovitz, M. A. (2017). Women
with Symptomatic Preterm Birth Have a Distinct Cervicovaginal Metabolome.
Am. J. Perinatol. 34, 10781083. doi: 10.1055/s-0037-1603817
Goldenberg, R. L., Hauth, J. C., and Andrews, W. W. (2000). Intrauterine infection
and preterm delivery. N.Engl.J.Med.342, 15001507. doi: 10.1056/
NEJM200005183422007
Goldenberg, R. L., Culhane, J. F., Iams, J. D., and Romero, R. (2008). Epidemiology
and causes of preterm birth. Lancet 371, 7584. doi: 10.1016/S0140-6736(08)
60074-4
Gong, Z., Luna, Y., Yu, P., and Fan, H. (2014). Lactobacilli inactivate Chlamydia
trachomatis through lactic acid but not H2O2. PloS One 9, e107758. doi:
10.1371/journal.pone.0107758
Haahr, T., Jensen, J. S., Thomsen, L., Duus, L., Rygaard, K., and Humaidan, P.
(2016). Abnormal vaginal microbiota may be associated with poor
reproductive outcomes: a prospective study in IVF patients. Hum. Reprod.
31, 795803. doi: 10.1093/humrep/dew026
Han, C., Li, H., Han, L., Wang, C., Yan, Y., Qi, W., et al. (2019). Aerobic vaginitis
in late pregnancy and outcomes of pregnancy. Eur. J. Clin. Microbiol. Infect.
Dis. 38, 233239. doi: 10.1007/s10096-018-3416-2
Harper, A., Pogson, C. I., Jones, M. L., and Pearce, J. H. (2000). Chlamydial
development is adversely affected by minor changes in amino acid supply,
blood plasma amino acid levels, and glucose deprivation. Infect. Immun. 68,
14571464. doi: 10.1128/IAI.68.3.1457-1464.2000
Hernandez-Vargas, P., Munoz, M., and Dominguez, F. (2020). Identifying
biomarkers for predicting successful embryo implantation: applying single to
multi-OMICs to improve reproductive outcomes. Hum. Reprod. Update 26,
264301. doi: 10.1093/humupd/dmz042
Hooper, L. V., Littman, D. R., and Macpherson, A. J. (2012). Interactions between
the microbiota and the immune system. Science 336, 12681273. doi: 10.1126/
science.1223490
Hopkins, M. M., and Meier, K. E. (2017). Free fatty acid receptor (FFAR) agonists
inhibit proliferation of human ovarian cancer cells. Prostaglandins Leukot.
Essent. Fatty Acids 122, 2429. doi: 10.1016/j.plefa.2017.06.013
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948817
Hyman, R. W., Herndon, C. N., Jiang, H., Palm, C., Fukushima, M., Bernstein, D.,
et al. (2012). The dynamics of the vaginal microbiome during infertility therapy
with in vitro fertilization-embryo transfer. J. Assist. Reprod. Genet. 29, 105115.
doi: 10.1007/s10815-011-9694-6
Icard, P., Shulman, S., Farhat, D., Steyaert, J. M., Alifano, M., and Lincet, H.
(2018). How the Warburg effect supports aggressiveness and drug resistance of
cancer cells? Drug Resist. Update 38, 111. doi: 10.1016/j.drup.2018.03.001
Idahl, A., Le Cornet, C., Gonzalez Maldonado, S., Waterboer, T., Bender, N.,
Tjønneland, A., et al. (2020). Serologic markers of Chlamydia trachomatis and
other sexually transmitted infections and subsequent ovarian cancer risk:
Results from the EPIC cohort. Int. J. Cancer 147, 20422052. doi: 10.1002/
ijc.32999
Ilhan,Z.E.,Laniewski,P.,Thomas,N.,Roe,D.J.,Chase,D.M.,andHerbst-Kralovetz,
M. M. (2019). Deciphering the complex interplay between microbiota, HPV,
inammation and cancer through cervicovaginal metabolic proling.
EBioMedicine 44, 675690. doi: 10.1016/j.ebiom.2019.04.028
Ismail, A. Q. T. (2018). Does placental MDSC-mediated modulation of arginine
levels help protect the foetus from auxotrophic pathogens? J. Matern. Fetal
Neonatal Med. 31, 16671669. doi: 10.1080/14767058.2017.1319935
Jasper, M. J., Tremellen, K. P., and Robertson, S. A. (2006). Primary unexplained
infertility is associated with reduced expression of the T-regulatory cell
transcription factor Foxp3 in endometrial tissue. Mol. Hum. Reprod. 12,
301308. doi: 10.1093/molehr/gal032
Johnson, R. M. (2004). Murine oviduct epithelial cell cytokine responses to
Chlamydia muridarum infection include interleukin-12-p70 secretion. Infect.
Immun. 72, 39513960. doi: 10.1128/IAI.72.7.3951-3960.2004
Jones, M. L., Ganopolsky, J. G., Labbe, A., Wahl, C., and Prakash, S. (2010).
Antimicrobial properties of nitric oxide and its application in antimicrobial
formulations and medical devices. Appl. Microbiol. Biotechnol. 88, 401407.
doi: 10.1007/s00253-010-2733-x
Kassie, F., Rabot, S., Kundi, M., Chabicovsky, M., Qin, H. M., and Knasmüller, S.
(2001). Intestinal microora plays a crucial role in the genotoxicity of the
cooked food mutagen 2-amino-3-methylimidazo [4,5-f]quinoline.
Carcinogenesis 22, 17211725. doi: 10.1093/carcin/22.10.1721
Klein, C., Gonzalez, D., Samwel, K., Kahesa, C., Mwaiselage, J., Aluthge, N., et al.
(2019). Relationship between the Cervical Microbiome, HIV Status, and
Precancerous Lesions. mBio 10, e02785-18. doi: 10.1128/mBio.02785-18
Koedooder, R., Mackens, S., Budding, A., Fares, D., Blockeel, C., Laven, J., et al.
(2019). Identication and evaluation of the microbiome in the female and male
reproductive tracts. Hum. Reprod. Update 25, 298325. doi: 10.1093/humupd/
dmy048
Kropf, P., Baud, D., Marshall, S. E., Munder, M., Mosley, A., Fuentes, J. M., et al.
(2007). Arginase activity mediates reversible T cell hyporesponsiveness in
human pregnancy. Eur. J. Immunol. 37, 935945. doi: 10.1002/eji.200636542
Kwon, M., Seo, S. S., Kim, M. K., Lee, D. O., and Lim, M. C. (2019). Compositional
and Functional Differences between Microbiota and Cervical Carcinogenesis as
Identied by Shotgun Metagenomic Sequencing. Cancers (Basel) 11, 309. doi:
10.3390/cancers11030309
Kyongo, J. K., Jespers, V., Goovaerts, O., Michiels, J., Menten, J., Fichorova, R. N.,
et al. (2012). Searching for lower female genital tract soluble and cellular
biomarkers: dening levels and predictors in a cohort of healthy Caucasian
women. PloS One 7, e43951. doi: 10.1371/journal.pone.0043951
Laniewski, P., Barnes, D., Goulder, A., Cui, H., Roe, D. J., Chase, D. M., et al.
(2018). Linking cervicovaginal immune signatures, HPV and microbiota
composition in cervical carcinogenesis in non-Hispanic and Hispanic
women. Sci. Rep. 8, 7593. doi: 10.1038/s41598-018-25879-7
Laniewski, P., Ilhan, Z. E., and Herbst-Kralovetz, M. M. (2020). The microbiome
and gynaecological cancer development, prevention and therapy. Nat. Rev.
Urol. 17, 232250. doi: 10.1038/s41585-020-0286-z
Leiby, J. S., Mccormick, K., Sherrill-Mix, S., Clarke, E. L., Kessler, L. R., Taylor,
L. J., et al. (2018). Lack of detection of a human placenta microbiome in
samples from preterm and term deliveries. Microbiome 6, 196. doi: 10.1186/
s40168-018-0575-4
Lewis, M. E., Belland, R. J., Abdelrahman, Y. M., Beatty, W. L., Aiyar, A. A., Zea, A.
H., et al. (2014). Morphologic and molecular evaluation of Chlamydia
trachomatis growth in human endocervix reveals distinct growth patterns.
Front. Cell Infect. Microbiol. 4, 71. doi: 10.3389/fcimb.2014.00071
Lim, E. S., Rodriguez, C., and Holtz, L. R. (2018). Amniotic uid from healthy term
pregnancies does not harbor a detectable microbial community. Microbiome 6,
87. doi: 10.1186/s40168-018-0475-7
Liu, L., Oza, S., Hogan, D., Chu, Y., Perin, J., Zhu, J., et al. (2016). Global, regional,
and national causes of under-5 mortality in 2000-15: an updated systematic
analysis with implications for the Sustainable Development Goals. Lancet 388,
30273035. doi: 10.1016/S0140-6736(16)31593-8
Liu, S., Wei, H., Li, Y., Huang, C., Lian, R., Xu, J., et al. (2018). Downregulation of
ILT4(+) dendritic cells in recurrent miscarriage and recurrent implantation
failure. Am. J. Reprod. Immunol. 80, e12998. doi: 10.1111/aji.12998
Ma, Y., Madupu, R., Karaoz, U., Nossa, C. W., Yang, L., Yooseph, S., et al. (2014).
Human papillomavirus community in healthy persons, dened by
metagenomics analysis of human microbiome project shotgun sequencing
data sets. J. Virol. 88, 47864797. doi: 10.1128/JVI.00093-14
MacIntyre, D. A., Chandiramani, M., Lee, Y. S., Kindinger, L., Smith, A.,
Angelopoulos, N., et al. (2015). The vaginal microbiome during pregnancy
and the postpartum period in a European population. Sci. Rep. 5, 8988. doi:
10.1038/srep08988
Macklaim, J. M., Fernandes, A. D., Di Bella, J. M., Hammond, J. A., Reid, G., and
Gloor, G. B. (2013). Comparative meta-RNA-seq of the vaginal microbiota and
differential expression by Lactobacillus iners in health and dysbiosis.
Microbiome 1, 12. doi: 10.1186/2049-2618-1-12
McMillan, A., Rulisa, S., Sumarah, M., Macklaim, J. M., Renaud, J., Bisanz, J. E.,
et al. (2015). A multi-platform metabolomics approach identies highly
specic biomarkers of bacterial diversity in the vagina of pregnant and non-
pregnant women. Sci. Rep. 5, 14174. doi: 10.1038/srep14174
Mei,C.,Yang,W.,Wei,X.,Wu,K.,andHuang,D.(2019).TheUnique
Microbiome and Innate Immunity During Pregnancy. Front. Immunol. 10,
2886. doi: 10.3389/mmu.2019.02886
Menon, R., Jones, J., Gunst, P. R., Kacerovsky, M., Fortunato, S. J., Saade, G. R.,
et al. (2014). Amniotic uid metabolomic analysis in spontaneous preterm
birth. Reprod. Sci. 21, 791803. doi: 10.1177/1933719113518987
Mirmonsef, P., Gilbert, D., Zariffard, M. R., Hamaker, B. R., Kaur, A., Landay,
A. L., et al. (2011). The effects of commensal bacteria on innate immune
responses in the female genital tract. Am. J. Reprod. Immunol. 65, 190195. doi:
10.1111/j.1600-0897.2010.00943.x
Mitchell, C., Manhart, L. E., Thomas, K., Fiedler, T., Fredricks, D. N., and
Marrazzo, J. (2012). Behavioral predictors of colonization with Lactobacillus
crispatus or Lactobacillus jensenii after treatment for bacterial vaginosis: a
cohort study. Infect. Dis. Obstet. Gynecol. 2012, 706540. doi: 10.1155/2012/
706540
Mitra, A., Macintyre, D. A., Lee, Y. S., Smith, A., Marchesi, J. R., Lehne, B., et al.
(2015). Cervical intraepithelial neoplasia disease progression is associated with
increased vaginal microbiome diversity. Sci. Rep. 5, 16865. doi: 10.1038/
srep16865
Molenaar, M. C., Singer, M., and Ouburg, S. (2018). The two-sided role of the
vaginal microbiome in Chlamydia trachomatis and Mycoplasma genitalium
pathogenesis. J. Reprod. Immunol. 130, 1117. doi: 10.1016/j.jri.2018.08.006
Monteiro, C., Marques, P. I., Cavadas, B., Damião, I., Almeida, V., Barros, N., et al.
(2018). Characterization of microbiota in male infertility cases uncovers
differences in seminal hyperviscosity and oligoasthenoteratozoospermia
possibly correlated with increased prevalence of infectious bacteria. Am. J.
Reprod. Immunol. 79, e12838. doi: 10.1111/aji.12838
Moreno, I., Codoner, F. M., Vilella, F., Valbuena, D., Martinez-Blanch, J. F.,
Jimenez-Almazan, J., et al. (2016). Evidence that the endometrial microbiota
has an effect on implantation success or failure. Am. J. Obstet. Gynecol. 215,
684703. doi: 10.1016/j.ajog.2016.09.075
Moreno, I., Garcia-Grau, I., Bau, D., Perez-Villaroya, D., Gonzalez-Monfort, M.,
Vilella, F., et al. (2020). The rst glimpse of the endometrial microbiota in early
pregnancy. Am.J.Obstet.Gynecol.222, 296305. doi: 10.1016/
j.ajog.2020.01.031
Nene, N. R., Reisel, D., Leimbach, A., Franchi, D., Jones, A., Evans, I., et al. (2019).
Association between the cervicovaginal microbiome, BRCA1 mutation status,
and risk of ovarian cancer: a case-control study. Lancet Oncol. 20, 11711182.
doi: 10.1016/S1470-2045(19)30340-7
Ness, R. B., Kip, K. E., Hillier, S. L., Soper, D. E., Stamm, C. A., Sweet, R. L., et al.
(2005). A cluster analysis of bacterial vaginosis-associated microora and
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948818
pelvic inammatory disease. Am. J. Epidemiol. 162, 585590. doi: 10.1093/aje/
kwi243
Nuriel-Ohayon, M., Neuman, H., and Koren, O. (2016). Microbial Changes during
Pregnancy, Birth, and Infancy. Front. Microbiol. 7, 1031. doi: 10.3389/
fmicb.2016.01031
Olive, A. J., and Sassetti, C. M. (2016). Metabolic crosstalk between host and
pathogen: sensing, adapting and competing. Nat. Rev. Microbiol. 14, 221234.
doi: 10.1038/nrmicro.2016.12
Organization,W.H.(2020).World Health Organization Global adult estimates of
chlamydia, gonorrhoea, trichomoniasis and syphilis including maternal and
congenital syphilis, 2016. Available at: https://www.who.int/reproductivehealth/
topics/rtis/STIs-Estimates.pdf?ua=1 (Accessed 2020).
Parolin, C., Foschi, C., Laghi, L., Zhu, C., Banzola, N., Gaspari, V., et al. (2018).
Insights Into Vaginal Bacterial Communities and Metabolic Proles of
Chlamydia trachomatis Infection: Positioning Between Eubiosis and
Dysbiosis. Front. Microbiol. 9, 600. doi: 10.3389/fmicb.2018.00600
Peebles, K., Velloza, J., Balkus, J. E., Mcclelland, R. S., and Barnabas, R. V. (2019). High
Global Burden and Costs of Bacterial Vaginosis: A Systematic Review and Meta-
Analysis. Sex Transm. Dis. 46, 304311. doi: 10.1097/OLQ.0000000000000972
Pelzer, E. S., Allan, J. A., Waterhouse, M. A., Ross, T., Beagley, K. W., and Knox,
C. L. (2013). Microorganisms within human follicular uid: effects on IVF. PloS
One 8, e59062. doi: 10.1371/journal.pone.0059062
Pereira, N., Edlind, T. D., Schlievert, P. M., and Nyirjesy, P. (2013). Vaginal toxic
shock reaction triggering desquamative inammatory vaginitis. J. Low. Genit.
Tract Dis. 17, 8891. doi: 10.1097/LGT.0b013e3182656991
Poisson, L. M., Munkarah, A., Madi, H., Datta, I., Hensley-Alford, S., Tebbe, C.,
et al. (2015). A metabolomic approach to identifying platinum resistance in
ovarian cancer. J. Ovarian Res. 8, 13. doi: 10.1186/s13048-015-0140-8
Postler, T. S., and Ghosh, S. (2017). Understanding the Holobiont: How Microbial
Metabolites Affect Human Health and Shape the Immune System. Cell Metab.
26, 110130. doi: 10.1016/j.cmet.2017.05.008
Prevention, C.F.D.C.A. (2019). Infertility Frequently Asked Questions [Online].
Centers for Disease Control and Prevention. Available at: https://www.cdc.gov/
reproductivehealth/infertility/index.htm (Accessed January 16 2019).
Prince, A. L., Ma, J., Kannan, P. S., Alvarez, M., Gisslen, T., Harris, R. A., et al.
(2016). The placental membrane microbiome is altered among subjects with
spontaneous preterm birth with and without chorioamnionitis. Am. J. Obstet.
Gynecol. 214, 627.e621627.e616. doi: 10.1016/j.ajog.2016.01.193
Pruski, P., Lewis, H. V., Lee, Y. S., Marchesi, J. R., Bennett, P. R., Takats, Z., et al.
(2018). Assessment of microbiota:host interactions at the vaginal mucosa
interface. Methods 149, 7484. doi: 10.1016/j.ymeth.2018.04.022
Rasmussen, S. J., Eckmann, L., Quayle, A. J., Shen, L., Zhang, Y. X., Anderson,
D. J., et al. (1997). Secretion of proinammatory cytokines by epithelial cells in
response to Chlamydia infection suggests a central role for epithelial cells in
chlamydial pathogenesis. J. Clin. Invest. 99, 7787. doi: 10.1172/JCI119136
Ravel, J., Gajer, P., Abdo, Z., Schneider, G. M., Koenig, S. S., Mcculle, S. L., et al.
(2011). Vaginal microbiome of reproductive-age women. Proc. Natl. Acad. Sci.
U.S.A. 108 Suppl 1, 46804687. doi: 10.1073/pnas.1002611107
Richardson, A. R., Dunman, P. M., and Fang, F. C. (2006). The nitrosative stress
response of Staphylococcus aureus is required for resistance to innate
immunity. Mol. Microbiol. 61, 927939. doi: 10.1111/j.1365-2958.2006.05290.x
Richardson, A. R., Libby, S. J., and Fang, F. C. (2008). A nitric oxide-inducible
lactate dehydrogenase enables Staphylococcus aureus to resist innate
immunity. Science 319, 16721676. doi: 10.1126/science.1155207
Rodriguez-Esquivel, M., Rosales, J., Castro, R., Apresa-Garcia, T., Garay, O.,
Romero-Morelos, P., et al. (2018). Volatolome of the Female Genitourinary
Area: Toward the Metabolome of Cervical Cancer. Arch. Med. Res. 49, 2735.
doi: 10.1016/j.arcmed.2018.04.004
Romanik,M.,Martirosian,G.,Wojciechowska-Wieja,A.,Cies
lik, K., and
Kazmierczak, W. (2007). [Co-occurence of indol-producing bacterial strains in the
vagina of women infected with Chlamydia trachomatis]. Ginekol. Pol. 78, 611615.
Romero, R., Hassan, S. S., Gajer, P., Tarca, A. L., Fadrosh, D. W., Nikita, L., et al.
(2014). The composition and stability of the vaginal microbiota of normal
pregnant women is different from that of non-pregnant women. Microbiome 2,
4. doi: 10.1186/2049-2618-2-4
Roth, A., König, P., Van Zandbergen, G., Klinger, M., Hellwig-Bürgel, T.,
Däubener, W., et al. (2010). Hypoxia abrogates antichlamydial properties of
IFN-gin human fallopian tube cells in vitro and ex vivo. Proc. Natl. Acad. Sci.
U.S.A. 107, 1950219507. doi: 10.1073/pnas.1008178107
Rotstein, O. D., Pruett, T. L., Fiegel, V. D., Nelson, R. D., and Simmons, R. L.
(1985). Succinic acid, a metabolic by-product of Bacteroides species, inhibits
polymorphonuclear leukocyte function. Infect. Immun. 48, 402408. doi:
10.1128/IAI.48.2.402-408.1985
Sasaki-Imamura,T.,Yoshida,Y.,Suwabe,K.,Yoshimura,F.,andKato,H.(2011).
Molecular basis of indole production catalyzed by tryptophanase in the genus
Prevotella. FEMS Microbiol. Lett. 322, 5159. doi: 10.1111/j.1574-6968.2011.02329.x
Scott, A. J., Alexander, J. L., Merrield, C. A., Cunningham, D., Jobin, C., Brown,
R., et al. (2019). International Cancer Microbiome Consortium consensus
statement on the role of the human microbiome in carcinogenesis. Gut 68,
16241632. doi: 10.1136/gutjnl-2019-318556
Serrano,M.G.,Parikh,H.I.,Brooks,J.P.,Edwards, D. J., Arodz, T. J., Edupuganti, L.,
et al. (2019). Racioethnic diversity in the dynamics of the vaginal microbiome
during pregnancy. Nat. Med. 25, 10011011. doi: 10.1038/s41591-019-0465-8
Sevin, D. C., Kuehne, A., Zamboni, N., and Sauer, U. (2015). Biological insights
through nontargeted metabolomics. Curr. Opin. Biotechnol. 34, 18. doi:
10.1016/j.copbio.2014.10.001
Shanmughapriya, S., Senthilkumar, G., Vinodhini, K., Das, B. C., Vasanthi, N., and
Natarajaseenivasan, K. (2012). Viral and bacterial aetiologies of epithelial
ovarian cancer. Eur. J. Clin. Microbiol. Infect. Dis. 31, 23112317. doi:
10.1007/s10096-012-1570-5
Shemer, Y., and Sarov, I. (1985). Inhibition of growth of Chlamydia trachomatis
by human gamma interferon. Infect. Immun. 48, 592596. doi: 10.1128/
IAI.48.2.592-596.1985
Sherrard, J., Wilson, J., Donders, G., Mendling, W., and Jensen, J. S. (2018). 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 29, 12581272. doi:
10.1177/0956462418785451
Shipitsyna, E., Khusnutdinova, T., Budilovskaya, O., Krysanova, A., Shalepo, K.,
Savicheva, A., et al. (2020). Bacterial vaginosis-associated vaginal microbiota is
an age-independent risk factor for Chlamydia trachomatis, Mycoplasma
genitalium and Trichomonas vaginalis infections in low-risk women, St.
Petersburg, Russia. Eur. J. Clin. Microbiol. Infect. Dis. 39, 12211230. doi:
10.1007/s10096-020-03831-w
Sierra-Filardi, E., Nieto, C., Domı
nguez-Soto, A., Barroso, R., Sanchez-Mateos, P.,
Puig-Kroger, A., et al. (2014). CCL2 shapes macrophage polarization by GM-
CSF and M-CSF: identication of CCL2/CCR2-dependent gene expression
prole. J. Immunol. 192, 38583867. doi: 10.4049/jimmunol.1302821
Sirota, I., Zarek, S. M., and Segars, J. H. (2014). Potential inuence of the
microbiome on infertility and assisted reproductive technology. Semin.
Reprod. Med. 32, 3542. doi: 10.1055/s-0033-1361821
Smith, S. B., and Ravel, J. (2017). The vaginal microbiota, host defence and
reproductive physiology. J. Physiol. 595, 451463. doi: 10.1113/JP271694
Snyers, D., Lefebvre, C., Viellevoye, R., and Rigo, V. (2020). [Late preterm : high
risk newborns despite appearances]. Rev. Med. Liege 75, 105110.
Son, K. A., Kim, M., Kim, Y. M., Kim, S. H., Choi, S. J., Oh, S. Y., et al. (2018).
Prevalence of vaginal microorganisms among pregnant women according to
trimester and association with preterm birth. Obstet. Gynecol. Sci. 61, 3847.
doi: 10.5468/ogs.2018.61.1.38
Song, J., Wang, X., Guo, Y., Yang, Y., Xu, K., Wang, T., et al. (2019). Novel high-
coverage targeted metabolomics method (SWATHtoMRM) for exploring
follicular uid metabolome alterations in women with recurrent spontaneous
abortion undergoing in vitro fertilization. Sci. Rep. 9, 10873. doi: 10.1038/
s41598-019-47370-7
Song, S. D., Acharya, K. D., Zhu, J. E., Deveney, C. M., Walther-Antonio, M. R. S.,
Tetel, M. J., et al. (2020). Daily Vaginal Microbiota Fluctuations Associated
with Natural Hormonal Cycle, Contraceptives, Diet, and Exercise. mSphere 5,
e0059320. doi: 10.1128/mSphere.00593-20
Spiegel, C. A., Amsel, R., Eschenbach, D., Schoenknecht, F., and Holmes, K. K.
(1980). Anaerobic bacteria in nonspecic vaginitis. N. Engl. J. Med. 303, 601
607. doi: 10.1056/NEJM198009113031102
Srinivasan, S., Morgan, M. T., Fiedler, T. L., Djukovic, D., Hoffman, N. G., Raftery,
D., et al. (2015). Metabolic signatures of bacterial vaginosis. mBio 6, e00204-15.
doi: 10.1128/mBio.00204-15
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948819
Stanek, R., Gain, R. E., Glover, D. D., and Larsen, B. (1992). High performance ion
exclusion chromatographic characterization of the vaginal organic acids in
women with bacterial vaginosis. BioMed. Chromatogr. 6, 231235. doi:
10.1002/bmc.1130060506
Strauss, J. F., Romero, R., Gomez-Lopez, N., Haymond-Thornburg, H., Modi,
B. P., Teves, M. E., et al. (2018). Spontaneous preterm birth: advances toward
the discovery of genetic predisposition. Am. J. Obstet. Gynecol. 218, 294
314.e292. doi: 10.1016/j.ajog.2017.12.009
Svenstrup, H. F., Fedder, J., Abraham-Peskir, J., Birkelund, S., and Christiansen, G.
(2003). Mycoplasma genitalium attaches to human spermatozoa. Hum.
Reprod. 18, 21032109. doi: 10.1093/humrep/deg392
Swiecki, M., Miller, H. L., Sesti-Costa, R., Cella, M., Gilllan, S., and Colonna, M.
(2017). Microbiota induces tonic CCL2 systemic levels that control pDC
trafcking in steady state. Mucosal Immunol. 10, 936945. doi: 10.1038/
mi.2016.99
Szewczyk, G., Maciejewski, T. M., and Szukiewicz, D. (2019). Current progress in
the inammatory background of angiogenesis in gynecological cancers.
Inammation Res. 68, 247260. doi: 10.1007/s00011-019-01215-1
Tao, Z., Zhang, L., Zhang, Q., Lv, T., Chen, R., Wang, L., et al. (2019). The
Pathogenesis Of Streptococcus anginosus In Aerobic Vaginitis. Infect. Drug
Resist. 12, 37453754. doi: 10.2147/IDR.S227883
Tavares-Murta, B. M., De Resende, A. D., Cunha, F. Q., and Murta, E. F. (2008).
Local prole of cytokines and nitric oxide in patients with bacterial vaginosis
and cervical intraepithelial neoplasia. Eur. J. Obstet. Gynecol. Reprod. Biol. 138,
9399. doi: 10.1016/j.ejogrb.2007.06.015
Troisi, J., Sarno, L., Landol, A., Scala, G., Martinelli, P., Venturella, R., et al.
(2018). Metabolomic Signature of Endometrial Cancer. J. Proteome Res. 17,
804812. doi: 10.1021/acs.jproteome.7b00503
Turkoglu, O., Zeb, A., Graham, S., Szyperski, T., Szender, J. B., Odunsi, K., et al. (2016).
Metabolomics of biomarker discovery in ovarian cancer: a systematic review of the
current literature. Metabolomics 12, 60. doi: 10.1007/s11306-016-0990-0
Van Der Veer,C., Hertzberger, R. Y., Bruisten,S. M., Tytgat, H. L. P., Swanenburg,J.,
De Kat Angelino-Bart, A., et al. (2019). Comparative genomics of human
Lactobacillus crispatus isolates reveals genes for glycosylation and glycogen
degradation: implications for in vivo dominance of the vaginal microbiota.
Microbiome 7, 49. doi: 10.1186/s40168-019-0667-9
VanHoudt,R.,Ma,B.,Bruisten,S.M.,Speksnijder,A.,Ravel,J.,andDeVries,H.J.C.
(2018). Lactobacillus iners-dominated vaginal microbiota is associated with increased
susceptibility to Chlamydia trachomatis infection in Dutch women: a case-control
study. Sex Transm. Infect. 94, 117123. doi: 10.1136/sextrans-2017-053133
Van Ostade, X., Dom, M., Tjalma, W., and Van Raemdonck, G. (2018). Candidate
biomarkers in the cervical vaginal uid for the (self-)diagnosis of cervical precancer.
Arch. Gynecol. Obstet. 297, 295311. doi: 10.1007/s00404-017-4587-2
Ventriglia, J., Paciolla, I., Pisano, C., Cecere, S. C., Di Napoli, M., Tambaro, R.,
et al. (2017). Immunotherapy in ovarian, endometrial and cervical cancer: State
of the art and future perspectives. Cancer Treat Rev. 59, 109116. doi: 10.1016/
j.ctrv.2017.07.008
Verstraelen, H., Vilchez-Vargas, R., Desimpel, F., Jauregui, R., Vankeirsbilck, N.,
Weyers, S., et al. (2016). Characterisation of the human uterine microbiome in
non-pregnant women through deep sequencing of the V1-2 region of the 16S
rRNA gene. PeerJ 4, e1602. doi: 10.7717/peerj.1602
Vick, E. J., Park, H. S., Huff, K. A., Brooks, K. M., Farone, A. L., and Farone, M. B.
(2014). Gardnerella vaginalis triggers NLRP3 inammasome recruitment in
THP-1 monocytes. J. Reprod. Immunol. 106, 6775. doi: 10.1016/
j.jri.2014.08.005
Vitali, B., Cruciani, F., Picone, G., Parolin, C., Donders, G., and Laghi, L. (2015).
Vaginal microbiome and metabolome highlight specic signatures of bacterial
vaginosis. Eur. J. Clin. Microbiol. Infect. Dis. 34, 23672376. doi: 10.1007/
s10096-015-2490-y
Vitko, N. P., Spahich, N. A., and Richardson, A. R. (2015). Glycolytic dependency
of high-level nitric oxide resistance and virulence in Staphylococcus aureus.
mBio 6, e00045-15. doi: 10.1128/mBio.00045-15
Walther-Antonio,M.R.,Chen,J.,Multinu,F.,Hokenstad,A.,Distad,T.J.,Cheek,E.H.,
et al. (2016). Potential contribution of the uterine microbiome in the development of
endometrial cancer. Genome Med. 8, 122. doi: 10.1186/s13073-016-0368-y
Wang, Q., Wurtz, P., Auro, K., Makinen, V. P., Kangas, A. J., Soininen, P., et al.
(2016). Metabolic proling of pregnancy: cross-sectional and longitudinal
evidence. BMC Med. 14, 205. doi: 10.1186/s12916-016-0733-0
Wang, C., Fan, A., Li, H., Yan, Y., Qi, W., Wang, Y., et al. (2020). Vaginal bacterial
proles of aerobic vaginitis: a case-control study. Diagn. Microbiol. Infect. Dis.
96, 114981. doi: 10.1016/j.diagmicrobio.2019.114981
Watson, E., and Reid, G. (2018). Metabolomics as a clinical testing method for the
diagnosis of vaginal dysbiosis. Am. J. Reprod. Immunol. 80, e12979. doi:
10.1111/aji.12979
Wee, B. A., Thomas, M., Sweeney, E. L., Frentiu, F. D., Samios, M., Ravel, J., et al.
(2018). A retrospective pilot study to determine whether the reproductive tract
microbiota differs between women with a history of infertility and fertile
women. Aust. N. Z. J. Obstet. Gynaecol. 58, 341348. doi: 10.1111/ajo.12754
Witkin, S. S., Kligman, I., Grifo, J. A., and Rosenwaks, Z. (1995). Chlamydia
trachomatis detected by polymerase chain reaction in cervices of culture-
negative women correlates with adverse in vitro fertilization outcome. J. Infect.
Dis. 171, 16571659. doi: 10.1093/infdis/171.6.1657
Witkin, S. S., Mendes-Soares, H., Linhares, I. M., Jayaram, A., Ledger, W. J., and
Forney, L. J. (2013). Inuence 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 4,
e00460-13. doi: 10.1128/mBio.00460-13
Wolrath, H., Boren, H., Hallen, A., and Forsum, U. (2002). Trimethylamine
content in vaginal secretion and its relation to bacterial vaginosis. Apmis 110,
819824. doi: 10.1034/j.1600-0463.2002.1101108.x
Wood, H., Fehlner-Gardner, C., Berry, J., Fischer, E., Graham, B., Hackstadt, T.,
et al. (2003). Regulation of tryptophan synthase gene expression in Chlamydia
trachomatis. Mol. Microbiol. 49, 13471359. doi: 10.1046/j.1365-
2958.2003.03638.x
Workowski, K. A., and Bolan, G. A. (2015). Sexually transmitted diseases
treatment guidelines 2015. MMWR Recomm. Rep. 64, 1137.
Yang, T. K., Chung, C. J., Chung, S. D., Muo, C. H., Chang, C. H., and Huang, C. Y.
(2015). Risk of Endometrial Cancer in Women With Pelvic Inammatory
Disease: A Nationwide Population-Based Retrospective Cohort Study.
Medicine (Baltimore) 94, e1278. doi: 10.1097/MD.0000000000001278
Yarbrough, V. L., Winkle, S., and Herbst-Kralovetz, M. M. (2015). Antimicrobial
peptides in the female reproductive tract: a critical component of the mucosal
immune barrier with physiological and clinical implications. Hum. Reprod.
Update 21, 353377. doi: 10.1093/humupd/dmu065
Yeoman, C. J., Thomas, S. M., Miller, M. E., Ulanov, A. V., Torralba, M., Lucas, S.,
et al. (2013). A multi-omic systems-based approach reveals metabolic markers
of bacterial vaginosis and insight into the disease. PloS One 8, e56111. doi:
10.1371/journal.pone.0056111
Yoon, B. H., Oh, S. Y., Romero, R., Shim, S. S., Han, S. Y., Park, J. S., et al. (2001).
An elevated amniotic uid matrix metalloproteinase-8 level at the time of mid-
trimester genetic amniocentesis is a risk factor for spontaneous preterm
delivery. Am.J.Obstet.Gynecol.185, 11621167. doi: 10.1067/
mob.2001.117678
Zhang,T.,Wu,X.,Yin,M.,Fan,L.,Zhang,H.,Zhao,F.,etal.(2012).
Discrimination between malignant and benign ovarian tumors by plasma
metabolomic proling using ultra performance liquid chromatography/mass
spectrometry. Clin. Chim. Acta 413, 861868. doi: 10.1016/j.cca.2012.01.026
Ziklo, N., Huston, W. M., Hocking, J. S., and Timms, P. (2016a). Chlamydia
trachomatis Genital Tract Infections: When Host Immune Response and the
Microbiome Collide. Trends Microbiol. 24, 750765. doi: 10.1016/
j.tim.2016.05.007
Ziklo, N., Huston, W. M., Taing, K., Katouli, M., and Timms, P. (2016b). In vitro
rescue of genital strains of Chlamydia trachomatis from interferon-gand
tryptophan depletion with indole-positive, but not indole-negative Prevotella
spp. BMC Microbiol. 16, 286. doi: 10.1186/s12866-016-0903-4
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Copyright © 2020 Li, Zang, Wang, Li, Fan, Han and Xue. This is an open-access
article distributed under the terms of the Creative Commons Attribution License
(CC BY). The use,distribution or reproduction in other forums is permitted, provided
the original author(s) and the copyright owner(s) are credited and that the original
publicationin this journal iscited, in accordance with accepted academic practice. No
use, distribution or reproduction is permitted which does not comply with these terms.
Li et al. The Female Genital Tract Microenvironment
Frontiers in Cellular and Infection Microbiology | www.frontiersin.org December 2020 | Volume 10 | Article 60948820
... Vaginose bacteriana (VB) é uma condição vaginal associada com uma alteração na microbiota vaginal normal (Li et al., 2020;Wiesenfeld et al., 2003). ...
... Tais metabólitos são identificados como biomarcadores para inflamações e doenças ginecológicas, podendo ser utilizados para diagnóstico, prognóstico e gravidade da doença. Atualmente a metabolômica (estudo dos metabólitos) identifica as vias de produção de aminoácidos, carboidratos e lipídeos na microbiota vaginal e vem relacionando-as com fatores ambientais (Li et al., 2020). ...
... Diferentes metabólitos microbianos atuam de formas diversas a fim de manter a vaginalis e as vaginolisinas produzidas levariam a efeitos citotóxicos contra as células epiteliais vaginais (Dong et al., 2023;Li et al., 2020;Mirzaei et al., 2023). o que facilita a colonização por bactérias anaeróbias que podem levar a uma disbiose (Pybus;Onderdonk, 1999;Tachedjian et al., 2017). ...
Article
A microbiota do trato genital feminino é uma comunidade diversa de microrganismos que desempenha um papel crucial na saúde das mulheres, influenciando a função reprodutiva e a defesa contra patógenos. Sua composição em situações normais é repleta de bactérias do gênero Lactobacillus, como Lactobacillus crispatus e Lactobacillus iners, que auxiliam no pH vaginal ácido e produzem subprodutos metabólicos benéficos, competindo com microrganismos patogênicos. Alterações nessa microbiota, causadas por fatores ambientais, podem levar à disbiose e diversas condições clínicas. Compreender essa microbiota e sua relação com a bioquímica bacteriana é crucial para o diagnóstico e tratamento dessas doenças, assim como para o desenvolvimento de estratégias preventivas, retratando de forma fidedigna quais microrganismos então presentes e como eles se beneficiam metabolicamente em determinadas situações patogênicas. Este artigo explora o perfil bacteriano do trato genital feminino, destacando a influência da microbiota e da atividade bioquímica metabólica bacteriana na saúde feminina e os mecanismos que levam a doenças quando essa microbiota está desequilibrada. Apesar de não estar completamente elucidado esses mecanismos, entender o perfil dessa microbiota a nível metabólico pode ser promissor para fins diagnósticos e terapêuticos, a fim de se obter novas alternativas de diagnóstico e tratamentos personalizados para medicina.
... These changes may affect the microbial ecosystem within the vagina, heightening the risk of vaginitis. For women experiencing regular menstrual cycles, the vaginal pH generally falls within the range of 3.8 to 5.0 [10]. Research has shown that sperm acts as a significant alkalizing agent, swiftly neutralizing vaginal acidity (increasing the pH to over 6-7) for several hours post-coitus, which is crucial for spermatozoa's ability to traverse the female reproductive tract. ...
Article
Objectives: To examine the role of vaginal pH and basal body temperature (BBT) metrics in enhancing fertility and menstrual health across global and ethnic contexts. Setting: Analysis of global and ethnic variations in fertility and menstrual health. Intervention: A critical examination of non-invasive measures such as vaginal pH and BBT, along with factors like ethnicity, diet, lifestyle, and geographical location, in predicting ovulation and improving reproductive health outcomes. Main Outcome Measure: The potential of vaginal pH and BBT monitoring, supported by technology, in offering personalized reproductive health insights. Results & Conclusion: Variations in pH and BBT monitoring are observed across different ethnic groups, influenced by external factors. Findings suggest that integrating these metrics with educational and interventional programs can benefit women's psycho-social behaviors, quality of life, and emotional well-being. Further research is needed to validate these methods and explore their application in clinical settings, aiming to provide reliable, accessible tools for managing reproductive health with a personalized approach.
... Microbial environment, metabolites and immune system components coordinate in reproductive system homeostasis. 1 In the postpartum period, the female reproductive tract is highly vulnerable to bacteria, which can easily invade the uterine flora and cause various reproductive pathologies like metritis and endometritis. 2,3 Lipopolysaccharide (LPS), one of the main components of Gram-negative bacteria's outer membrane, can harm reproductive functions such as follicular and embryonic development, ovulation and implantation. ...
Article
Full-text available
This study investigated the protective effects of thymol on LPS-induced ovarian damage. Female Wistar albino rats were divided into five groups (n=35, group=7): control, solvent, LPS, thymol, and LPS+thymol. At the end of the study, hormone (E2 and AMH), oxidative stress (MDA, GSH, GSH-Px and catalase), and inflammatory cytokine (TNF-α and IL-6) analyses were performed in the samples taken from the animals. Ovary and body weights were also measured. Lipopolysaccharide treatment caused decreases in E2 AMH, GSH, GSH-Px and catalase levels but increased MDA, TNF-α and IL-6 levels. In the Lps+thymol group, thymol administration caused increases in E2, AMH, GSH, GSH-Px and catalase levels and decreased MDA, TNF-α and IL-6 levels. In conclusion, thymol administration positively affected Lps-induced hormonal, oxidative stress and inflammatory changes via antioxidant and anti-inflammatory mechanisms. However, it is thought that the long-term effects of thymol need to be demonstrated, especially by further molecular mechanisms.
Article
Full-text available
Microplastics have been detected in various human organs, and studies on their impact on human health are ongoing. However, few studies have researched microplastics in the cervicovaginal area. In this study, we aimed to assess their presence in human cervicovaginal lavage fluid. This prospective study was conducted at a single tertiary medical center, enrolling 10 participants aged 27–49 years. Human cervicovaginal lavage fluid samples were collected from the patients by a single skilled obstetrician. Raman spectroscopy was used to analyze and characterize microplastic particles detected in the samples. Ninety-one microplastic particles were detected in 10 samples. More than 50% of the microplastic particles were identified in a single patient who regularly used menstrual cups. The mean number of microplastics was 9.10 ± 14.96 per 10 g sample. Most of the microplastics were <50 μm in size, and polypropylene and polystyrene were the most predominant types. Raman analysis detected microplastic particles in human cervicovaginal lavage fluids, suggesting that the human cervicovaginal area is exposed to microplastics. The number of detected particles varied significantly among individuals. This study highlights the need for further research on the effects of microplastics on the female reproductive system using cervicovaginal lavage fluid.
Article
Full-text available
The vaginal microbiome of healthy women is dominated by Lactobacillus spp. A variety of illnesses, such as vaginosis, sexually transmitted infections (STIs), failed implantation, premature birth (PTB), and preterm pre-labor membrane rupture, are brought on by an unbalanced microbiota. Pregnancy is associated with a decrease in the metabolic capacity of the vaginal resident microbiome, which is consistent with a change to a less complex Lactobacillus-dominated microbiome. Age, race, sexual intercourse, smoking, IUD, contraception, lifestyle, and diet all affect the makeup of the vaginal microbiome. Moreover, physiological events including menarche, the menstrual cycle, pregnancy, menopause, and other hormonal changes have an impact on the vaginal microbiome. The vaginal microbiome is significantly disrupted by the menstrual cycle, with significant changes toward a more varied microbiota occurring around menstruation. Several major factors maintain or disrupt the vaginal microbiome including ethnic group, menstruation cycle, and pregnancy which are discussed in this section. In the index pregnancy, the vaginal microbiota of women who had already given birth, or had just experienced an induced or spontaneous abortion, was qualitatively and quantitatively different from that of women who were having their first child. Early pregnancy vaginal microbiome depletion is a risk factor for early pregnancy miscarriage. Although, early pregnancy miscarriage is not always caused by a high bacterial diversity and quantity of lactobacilli. Lactobacillus protects against pathogens through the production of antibacterial compounds such as lactic acid and bacteriocins.
Article
Full-text available
Previous studies showed that the female genital tract microbiome plays a crucial role in regulating the host’s immune defense mechanisms. Our previous research has shown that Lactobacillus gasseri LGV03 (L. gasseri LGV03) isolated from cervico-vagina of HPV-cleared women contributes to clearance of HPV infection and beneficially regulate immune response. However, the mechanisms behind the regulation of L. gasseri LGV03 in immune response remain unclear. To better understand the interaction between female genital tract microbiome and immune function, the immunomodulatory activities of L. gasseri LGV03 were investigated in zebrafish models of neutropenia, macrophage and T cells deficiency. L. gasseri LGV03 showed higher potent activities in ameliorating vinorelbine-induced neutropenia, macrophage and T cells deficiency, and significantly enhanced mRNA expressions of cytokines TNF-α, TNF-β and IFN-α. Moreover, the transcriptome sequencing results indicated L. gasseri LGV03 might alleviate vinorelbine-induced immunosuppression in zebrafish. Non-targeted detection and analysis revealed that indole derivatives including phenylacetaldehyde, 3-phenyllactic acid, N-acetylserotonin and indole-3-lactic acid were significantly increased in the lysate and supernatant of L. gasseri LGV03. Meanwhile, L. gasseri LGV03 supernatant and indole-3-lactic acid ameliorated the vinorelbine-induced reduction in abundance of macrophages, neutrophils and T cells. However, the alleviating effects of L. gasseri LGV03 supernatant or indole-3-lactic acid were eliminated by aryl hydrocarbon receptor (AHR) antagonist CH-223,191. Furthermore, L. gasseri LGV03 supernatant and indole-3-lactic acid significantly increased the secretion of IFN-α, IFN-β and chemokines (MIP-1α, MIP-1β) in Ect1/E6E7 cells, meanwhile, these benefits were eliminated by CH-223,191 treatment. In summary, L. gasseri LGV03-derived indole-3-lactic acid can activate AHR-mediated immune response.
Article
Full-text available
(1) Background: Microbiota could be related to tumorigenesis through the persistence of an inflammatory state, also at the endometrial level. Inflammation, in fact, is involved in the promotion of genetic instability and in a favorable microenvironment for tumor growth. One pathway could be the disruption of the epithelial/mucosal barrier, with the activation of cytokines. The microbiota also seem to favor other involved patterns, such as insulin resistance and increased adipose tissue. (2) Methods: The online search for this review was based on keywords such as “endometrial cancer” and “microbiota” on the main online scientific database. Our objective is a narrative up-to-date review of the current literature on gynecological microbiota; we analyze the possible correlations with known modifying and promoting oncological factors (i.e., Body Mass Index- BMI, menopause, pH), with particular attention to vaginal and uterine microorganisms respective to the development of endometrial cancer in comparison to healthy women. (3) Results: Various species and distributions of bacteria could be related to tumorigenesis and induce alterations in cell signaling and cycle pathways, including those in the gynecological field. (4) Conclusions: In the literature, the different composition of uterine and vaginal microbiota has been analyzed in the past years, and their diversity and actions seem to correlate with possible oncological effects.
Article
Full-text available
Endometritis is mostly caused by childbirth or postpartum uterine infection. It is one of the important reasons leading to female infertility. Caffeic acid (CA) and its derivatives are widely found in some foods and traditional Chinese medicine, and have biological activities such as antioxidant, free radical scavenging, anti‐inflammatory, and anti‐infection. In this study, we aimed to explore the effect of CA on Staphylococcus aureus‐induced endometritis. The contents of TNF‐α and IL‐1β were detected by ELISA in S. aureus‐induced endometritis model. Western blot assay was used to detect the expression of AMPKα/mTOR/HIF‐1α pathway related proteins and GPX4 expression. In addition, the concentrations of MDA, GSH, and iron were tested by the assay kits. Compared with the model group, CA treatment significantly alleviated S. aureus‐induced uterine injury, MPO activity, the contents of inflammatory factors TNF‐α and IL‐1β, and NF‐κB activation. Meanwhile, CA significantly inhibited S. aureus‐induced ferroptosis, as confirmed by decreased MDA and iron concentration and up‐regulated GPX4 expression and GSH level. Furthermore, CA attenuated S. aureus‐induced HIF‐1α and phosphorylated mTOR expression and increased phosphorylated AMPK expression. In conclusion, CA inhibits inflammation and ferroptosis by regulating AMPKα/mTOR/HIF‐1α signalling pathway to alleviate S. aureus‐induced endometritis in mice.
Article
Full-text available
Purpose Characterization of the human microbiome has become more precise with the application of powerful molecular tools utilizing the unique 16S ribosomal subunit’s hypervariable regions to greatly increase sensitivity. The microbiome of the lower genital tract can prognosticate obstetrical outcome while the upper reproductive tract remains poorly characterized. Here, the endometrial microbiome at the time of single embryo transfer (SET) is characterized by reproductive outcome. Methods Consecutive patients undergoing euploid, SET was included in the analysis. After embryo transfer, performed as per routine, the most distal 5-mm portion of the transfer catheter was sterilely placed in a DNA free PCR tube. Next-generation sequencing of the bacteria specific 16S ribosome gene was performed, allowing genus and species calls for microorganisms. Results Taxonomy assignments were made on 35 samples from 33 patients and 2 Escherichia coli controls. Of the 33 patients, 18 had ongoing pregnancies and 15 did not. There were a total of 278 different genus calls present across patient samples. The microbiome at time of transfer for those patients with ongoing pregnancy vs. those without ongoing pregnancy was characterized by top genera by sum fraction. Lactobacillus was the top species call for both outcomes. Conclusions The data presented here show the microbiome at the time of embryo transfer can successfully be characterized without altering standard clinical practice. This novel approach, both in specimen collection and analysis, is the first step toward the goal of determining physiologic from pathophysiologic microbiota. Further studies will help delineate if differences in the microbiome at the time of embryo transfer have a reliable impact on pregnancy outcome.
Article
Full-text available
The vaginal microbiome is a critical component of women’s sexual and reproductive health, with variations in microbial composition, particularly the loss of Lactobacillus species, being implicated in gynecologic and obstetric diseases. Given that the vaginal microbiome is so crucial, why do vaginal microbial profiles vary strikingly from person to person and even change over time within the same person? In the present study, which tracked the daily vaginal microbiomes of young healthy women through different lifestyles, we found that use of a locally released progestin contraceptive, a vegetarian diet, and intense exercise appear to lead to vaginal microbiome alterations and loss of Lactobacillus species. The impact of these vaginal microbiome changes on immediate and long-term health remain to be investigated.
Article
Full-text available
A substantial proportion of epithelial ovarian cancer (EOC) arises in the fallopian tube and other epithelia of the upper genital tract; these epithelia may incur damage and neoplastic transformation after sexually transmitted infections (STI) and pelvic inflammatory disease. We investigated the hypothesis that past STI infection, particularly Chlamydia trachomatis, is associated with higher EOC risk in a nested case‐control study within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort including 791 cases and 1669 matched controls. Serum antibodies against C. trachomatis, Mycoplasma genitalium, herpes simplex virus type 2 (HSV‐2) and human papillomavirus (HPV) 16, 18 and 45 were assessed using multiplex fluorescent bead‐based serology. Conditional logistic regression was used to estimate relative risks (RR) and 95% confidence intervals (CI) comparing women with positive vs. negative serology. A total of 40% of the study population was seropositive to at least one STI. Positive serology to C. trachomatis Pgp3 antibodies was not associated with EOC risk overall, but with higher risk of the mucinous histotype (RR = 2.30 [95% CI = 1.22‐4.32]). Positive serology for chlamydia heat shock protein 60 (cHSP60‐1) was associated with higher risk of EOC overall (1.36 [1.13‐1.64]) and with the serous subtype (1.44 [1.12‐1.85]). None of the other evaluated STIs were associated with EOC risk overall; however, HSV‐2 was associated with higher risk of endometrioid EOC (2.35 [1.24‐4.43]). The findings of our study suggest a potential role of C. trachomatis in the carcinogenesis of serous and mucinous EOC, while HSV‐2 might promote the development of endometrioid disease.
Article
Full-text available
Background: Successful embryo implantation is a complex process that requires the coordination of a series of events, involving both the embryo and the maternal endometrium. Key to this process is the intricate cascade of molecular mechanisms regulated by endocrine, paracrine and autocrine modulators of embryonic and maternal origin. Despite significant progress in ART, implantation failure still affects numerous infertile couples worldwide and fewer than 10% of embryos successfully implant. Improved selection of both the viable embryos and the optimal endometrial phenotype for transfer remains crucial to enhancing implantation chances. However, both classical morphological embryo selection and new strategies incorporated into clinical practice, such as embryonic genetic analysis, morphokinetics or ultrasound endometrial dating, remain insufficient to predict successful implantation. Additionally, no techniques are widely applied to analyse molecular signals involved in the embryo-uterine interaction. More reliable biological markers to predict embryo and uterine reproductive competence are needed to improve pregnancy outcomes. Recent years have seen a trend towards 'omics' methods, which enable the assessment of complete endometrial and embryonic molecular profiles during implantation. Omics have advanced our knowledge of the implantation process, identifying potential but rarely implemented biomarkers of successful implantation. Objective and rationale: Differences between the findings of published omics studies, and perhaps because embryonic and endometrial molecular signatures were often not investigated jointly, have prevented firm conclusions being reached. A timely review summarizing omics studies on the molecular determinants of human implantation in both the embryo and the endometrium will help facilitate integrative and reliable omics approaches to enhance ART outcomes. Search methods: In order to provide a comprehensive review of the literature published up to September 2019, Medline databases were searched using keywords pertaining to omics, including 'transcriptome', 'proteome', 'secretome', 'metabolome' and 'expression profiles', combined with terms related to implantation, such as 'endometrial receptivity', 'embryo viability' and 'embryo implantation'. No language restrictions were imposed. References from articles were also used for additional literature. Outcomes: Here we provide a complete summary of the major achievements in human implantation research supplied by omics approaches, highlighting their potential to improve reproductive outcomes while fully elucidating the implantation mechanism. The review highlights the existence of discrepancies among the postulated biomarkers from studies on embryo viability or endometrial receptivity, even using the same omic analysis. Wider implications: Despite the huge amount of biomarker information provided by omics, we still do not have enough evidence to link data from all omics with an implantation outcome. However, in the foreseeable future, application of minimally or non-invasive omics tools, together with a more integrative interpretation of uniformly collected data, will help to overcome the difficulties for clinical implementation of omics tools. Omics assays of the embryo and endometrium are being proposed or already being used as diagnostic tools for personalised single-embryo transfer in the most favourable endometrial environment, avoiding the risk of multiple pregnancies and ensuring better pregnancy rates.
Article
Full-text available
The large majority of studies investigating associations between bacterial vaginosis (BV) and sexually transmitted infections (STIs) have been conducted among predominantly young women with high risk for STIs. Since a risky sexual behavior is a significant risk factor for both STIs and BV, this creates a bias toward an increased association between BV and STIs. This study evaluated associations between BV-associated vaginal microbiota and STIs (Chlamydia trachomatis, Mycoplasma genitalium, Trichomonas vaginalis, and Neisseria gonorrhoeae) in a population of women with low risk for STIs and investigated STI outcomes depending on the dominating Lactobacillus species. Repository cervicovaginal samples collected from reproductive-age women from January 2014 to February 2019 were characterized for vaginal microbiota types and the STIs using multiplex real-time PCR assays. In total, 95 STI-positive and 91 STI-negative samples were included. A significant, age-independent association between BV-associated vaginal microbiota and the presence of C. trachomatis, M. genitalium, and T. vaginalis infections was identified (age-adjusted odds ratios 2.92 [95% confidence interval (CI) 1.24-7.03], 2.88 [95% CI 1.19-7.16], and 9.75 × 107 [95% CI 13.03-∞], respectively). Normal vaginal microbiota dominated by Lactobacillus crispatus, L. gasseri, or L. jensenii was a strong protective factor against C. trachomatis and/or M. genitalium infections, whereas L. iners-dominated microbiota was not significantly associated with C. trachomatis and/or M. genitalium positivity. The results of the present study confirm that STI prevention strategies should include interventions that also reduce the incidence of BV and promote a protective vaginal microbiota in both high- and low-risk women.
Article
Full-text available
In this study, we evaluate the association between vaginal and cervical human papillomavirus infections high-risk types (HPV+H), negative controls (HPV−), the bacterial biota, and urinary metabolites via integration of metagenomics, metabolomics, and bioinformatics analysis. We recently proposed that testing urine as a biofluid could be a non-invasive method for the detection of cervical HPV+H infections by evaluating the association between cervical HPV types and a total of 24 urinary metabolites identified in the samples. As a follow-up study, we expanded the analysis by pairing the urine metabolome data with vaginal and cervical microbiota in selected samples from 19 Puerto Rican women diagnosed with HPV+H infections and HPV− controls, using a novel comprehensive framework, Model-based Integration of Metabolite Observations and Species Abundances 2 (MIMOSA2). This approach enabled us to estimate the functional activities of the cervicovaginal microbiome associated with HPV+H infections. Our results suggest that HPV+H infections could induce changes in physicochemical properties of the genital tract through which niche partitioning may occur. As a result, Lactobacillus sp. enrichment coincided with the depletion of L. iners and Shuttleworthia, which dominate under normal physiological conditions. Changes in the diversity of microbial species in HPV+H groups influence the capacity of new community members to produce or consume metabolites. In particular, the functionalities of four metabolic enzymes were predicted to be associated with the microbiota, including acylphosphatase, prolyl aminopeptidase, prolyl-tRNA synthetase, and threonyl-tRNA synthetase. Such metabolic changes may influence systemic health effects in women at risk of developing cervical cancer. Overall, even assuming the limitation of the power due to the small sample number, our study adds to current knowledge by suggesting how microbial taxonomic and metabolic shifts induced by HPV infections may influence the maintenance of microbial homeostasis and indicate that HPV+H infections may alter the ecological balance of the cervicovaginal microbiota, resulting in higher bacterial diversity.
Article
Transcription profiles of genes of local immune response were determined in the vagina of women with bacterial vaginosis, aerobic vaginitis, and vulvovaginal candidosis for detection of the most specific immune markers for these vaginal infections. Laboratory diagnosis of the vaginal infections was performed microscopically; the inflammatory reaction in the vagina (leukorrhea) was defined as the presence of >10 white blood cells per field of view. Transcription profiles of IL1b, IL10, IL18, TNFα, TLR4, GATA3, and CD68 were determined using reverse-transcription quantitative real-time PCR. The strongest predictors of aerobic vaginitis were increased levels of IL1b and IL10 mRNA. Bacterial vaginosis was strongly associated with reduced levels of IL18 and GATA3 mRNA. Increased levels of IL1b and TLR4 transcripts showed significant discriminatory power for vulvovaginal candidosis and leukorrhea. The results of this study suggest differential expression of local immune response genes in the vagina of women with different vaginal infections. Detection of specific immune markers in the vagina using reverse-transcriptase PCR could supplement PCR detection of abnormal vaginal microflora for the diagnosis of vaginal infections.
Article
The female reproductive tract (FRT), similar to other mucosal sites, harbours a site-specific microbiome, which has an essential role in maintaining health and homeostasis. In the majority of women of reproductive age, the microbiota of the lower FRT (vagina and cervix) microenvironment is dominated by Lactobacillus species, which benefit the host through symbiotic relationships. By contrast, the upper FRT (uterus, Fallopian tubes and ovaries) might be sterile in healthy individuals or contain a low-biomass microbiome with a diverse mixture of microorganisms. When dysbiosis occurs, altered immune and metabolic signalling can affect hallmarks of cancer, including chronic inflammation, epithelial barrier breach, changes in cellular proliferation and apoptosis, genome instability, angiogenesis and metabolic dysregulation. These pathophysiological changes might lead to gynaecological cancer. Emerging evidence shows that genital dysbiosis and/or specific bacteria might have an active role in the development and/or progression and metastasis of gynaecological malignancies, such as cervical, endometrial and ovarian cancers, through direct and indirect mechanisms, including modulation of oestrogen metabolism. Cancer therapies might also alter microbiota at sites throughout the body. Reciprocally, microbiota composition can influence the efficacy and toxic effects of cancer therapies, as well as quality of life following cancer treatment. Modulation of the microbiome via probiotics or microbiota transplant might prove useful in improving responsiveness to cancer treatment and quality of life. Elucidating these complex host–microbiome interactions, including the crosstalk between distal and local sites, will translate into interventions for prevention, therapeutic efficacy and toxic effects to enhance health outcomes for women with gynaecological cancers.
Article
Investigation of the microbial community in the female reproductive tract using sequencing techniques has revealed that endometrial samples obtained through a transvaginal catheter are dominated by Lactobacillus species. Dysbiotic changes in the endometrial microbiota may be associated with implantation failure or early spontaneous abortion in patients undergoing assisted reproductive technology (ART) treatment. Whether or not there is an endometrial microbiota in early pregnancy is unknown. Herein we describe, the human endometrial microbiota in a patient who subsequently had an 8th week spontaneous clinical miscarriage with euploid embryos in the next cycle and, for the first time, during a successful pregnancy in which the endometrial fluid was sampled at 4 weeks of gestation. The microbial profile found on the endometrial sample prior to the spontaneous abortion had higher bacterial diversity and lower Lactobacillus abundance than the endometrial fluid from the healthy pregnancy. Functional metagenomics detected different Lactobacillus species between the two samples. Lactobacillus crispatus was present in the endometrium prior to the spontaneous abortion, as were other bacteria involved in dysbiosis, which had an unstable functional pattern characterized by transposases and insertion elements. Lactobacillus iners was the most prevalent microbe found in the endometrium during early pregnancy, associating its presence with defense mechanisms and basal functions. These novel observations prompt future investigations to understand the potential implications of microbiology on healthy and pathologic human pregnancy.
Article
Late preterm infants are born between 34 weeks of amenorrhea and 36 weeks 6 days. Late preterms represent the largest proportion of premature infants (about 75 %). Late prematurity is increasing in recent decades. While studies initially focused on mortality and morbidity related to very preterm birth, the late preterms have been the subject of increased attention over the past 15 years. Late preterm infants have an increased risk of respiratory complications, infections, feeding problems, hypothermia and hypoglycemia. Neonatal, infant and during adulthood mortalities are significantly higher in late preterm than in term infants. In addition, late preterm infants carry an increased risk of long-term morbidities, such as neurodevelopmental delay, cerebral palsy, chronic respiratory or metabolic diseases. This review highlights the evidence that late preterm infants are high risk newborns and require adapted follow-up.