1. Introduction
The
Lactobacillus group (
sensu lato) represented by over two hundred species and now classified into dozens of new genera [
1] is among the most widely distributed taxa on Earth. It is present in practically all viable environments in different climate zones and comes into close interactions with multicellular host organisms, ranging from plants to primates. This unusual versatility of
Lactobacillus bacteria is subsequent to its adaptability to various life conditions thanks to flexibility of their core metabolic pathways. These pathways include pyruvate metabolism, carbohydrate transport and metabolism, the proteolytic system, amino acid metabolism, and protein synthesis. This metabolic flexibility allows Lactobacilli to adjust protein levels in response to varying environmental conditions, enabling their survival and growth across diverse ecological niches [
2]. Moreover, beyond general stress responses and regulatory mechanisms, specific metabolic pathways can be selectively activated or deactivated, leading to changes in the behavior of the strains [
3]. One of the most well recognized
Lactobacillus species found in different fermentation products but also in the human gut microbiome, thanks to its capabilities of adaptation and genome plasticity is
L. (Lactiplantibacillus) plantarum [
4].
Research on probiotics, aimed at uncovering their functional mechanisms, provides much of the evidence emphasizing the critical role of the Lactobacillus group in the housekeeping of human health. Although extending these findings to all Lactobacillus strains that inhabit human body surfaces or diverse ecological niches is challenging, it is reasonable to speculate that the beneficial properties observed in specific probiotic strains might be broadly characteristic of the species. Given that probiotic lactobacilli were historically isolated at random from different niches within the healthy human microbiome and subsequently characterized, it is likely that many other Lactobacillus strains with similar properties exist in the human microbiome but have yet to be identified or studied.
The human gastrointestinal (GI) tract is home to a diverse and ever-changing array of microorganisms, including the gut microbiota, which play a crucial role in maintaining host homeostasis and regulating metabolic functions. Analysis of various metagenomic studies has revealed a total of 2,172 distinct microbial species present in humans, categorized into 12 phyla. Of these, a significant majority—93.5%—are classified under the Proteobacteria, Firmicutes, Actinobacteria, and Bacteroidetes phyla. Within this population, 386 species are known to be strictly anaerobic and are predominantly located in the GI tract [
5]. A comprehensive catalog of the gut microbiome’s functional potential has recently been created, identifying a total of 9,879,896 genes from a combination of 249 newly sequenced and 1,018 previously published samples [
6]. Despite not being the predominant group in the adult human gut microbiome, lactobacilli are essential for the early development of this microbiome.
It has been acknowledged for over a century that the gut microbiome plays a crucial role in health, a concept first explored through Metchnikoff’s groundbreaking research on lactic acid bacteria, particularly
Lactobacillus delbrueckii ssp. Bulgaricus [
7]. Recent years have brought an increasing number of data which causes appreciation of the infant gut microbiome and especially on the critical window of its highly dynamic maturation. Recent findings shed light on the significant influence of the gut microbiome on early life development, with a particular focus on how it affects the maturation of the immune system [
8]. While the precise molecular processes involved in neonatal priming in humans remain unclear, recent animal research indicates that the way infants acquire their gut microbiota is shaped by the interplay between bacterial and host factors. Therefore, the timing of bacterial colonization seems to be crucial for the development of the gut microbiome. For instance,
Lactobacillus species are abundant in the vaginal microbiota during the final stages of pregnancy and are also present in large numbers in human milk. Consequently, at birth, lactobacilli outnumber other genera, such as
Bifidobacterium, when colonizing the mucosal and skin surfaces of newborns. This initial colonization happens through exposure to the
Lactobacillus-rich environment of the vagina during delivery and is further supported by breastfeeding, which provides additional lactobacilli from the mother’s milk.
Colonization of the neonate surfaces with Firmicutes with predominance of lactobacilli, but also bifidobacteria and coagulase-negative staphylococci has also an immediate effect on the neonate innate immunity by activation of its different mechanisms, exemplified by stimulation of production of the tight-junction proteins by gut epithelial cells, resulting in gut impermeability for proteins and other macroparticles but also bacteria and thus preventing bacterial translocation from the gut lumen to lymphatics and bloodstream. In preterm neonates, especially those with low and very low birth weights, the lack of gut tightness often leads to neonatal sepsis [
9,
10,
11]. Current understanding of gut colonization suggests that the beneficial microbiota, transferred from the adult host to the neonate, is established during early infancy. This early microbial community has lasting effects, extending into adulthood and providing protection against systemic infections. Therefore, both the timing and selection of bacterial strains are critical for the effective artificial colonization of at-risk neonates. The success of this strategy is likely to improve as we identify these and other influential factors in humans and validate the outcomes of artificial colonization through clinical trials [
12].
2. Early Gut Colonization: Sources of Lactobacilli
There is more and more evidence showing that DNA from
Lactobacillus,
Bifidobacterium and other mostly anaerobic bacteria is found in most placenta samples from apparently healthy labors. There are also studies that have found living bacteria in the placenta and amniotic fluid, although their presence in these tissues seems to be related to chorioamnionitis and in utero inflammation [
13]. Thus, at the moment it is safer to accept that bacterial DNA is transferred horizontally from mother to fetus across the placenta under normal physiological conditions. Unmethylated CpG oligodeoxynucleotide motifs within bacterial DNA induce immune responses. Toll-like receptor 9 is activated by specific CpG motifs, which then trigger Th-1-type immune reactions. Even though newborns are generally viewed as immunologically underdeveloped, exposure to bacterial DNA might influence and shape their immune system during fetal development earlier than was once believed [
14]. Although there are studies indicating a presence of viable bacteria, but not lactobacilli, in amniotic fluid, evidence is not strong enough to accept that this is a physiological mechanism of maternal—fetal transfer of the bacteria [
15,
16].
From the moment of birth and in the hours that follow, through interactions with the external environment, such as breathing, breastfeeding, and contact with parents and healthcare providers, the baby’s mouth is exposed to numerous microbes. This exposure initiates the process of permanent colonization of the oral cavity during the postpartum period. Within just twenty-four hours, the first microorganisms, known as pioneer species, begin to establish themselves in the infant’s mouth. At this early stage, the oral cavity is most commonly colonized by Gram-positive cocci, particularly
Streptococcus,
Staphylococcus, and
Lactobacillus sensu lato 17,
18,].
In fact, literature on oral microbiota just after birth is based on studies done in the previous century, and what is even worse, using only qualitative cultures [
18]. Despite rather simple and non-accurate methods it was possible to demonstrate that bacterial strains obtained from the mother’s vagina and newborn’s mouth were identical [
19]. It seems that most of the organisms present in the newborn oral cavity represent only planktonic but not resident part of the microbiota and that some species which are extremely rarely found in a healthy vagina at birth may represent hospital pathogens. These bacteria do not form early gut microbiota found in meconium which is very similar to amniotic and vaginal microbiota. Using a modern sequencing approach, He et al. found various
Lactobacillus operational taxonomic units (OTUs) together with diverse strict anaerobic taxons OTUs but not those of
Staphylococcus [
20].
After birth, breast milk is crucial for establishing bacterial colonization in the infant’s gut. During the lactation period, the gut undergoes a progressive and sequential colonization by various microbial communities, transitioning from an aerobic, milk-focused microbiome in infancy to a more complex, adult-like microbiome as the child grows [
21]. Human milk is a primary source of bacteria for breastfed infants; between 10
5 and 10
7 bacteria are ingested daily by a baby consuming about 800 mL/day of milk. Notably, viable
Lactobacillus and
Bifidobacterium species are common in the milk of women who have not been treated with antibiotics, with their DNA detectable in most milk samples [
22]. Species such as
L. gasseri,
L. salivarius,
L. reuteri,
L. fermentum, and
B. breve are found to be more frequent among the bacteria isolated from human milk compared to others. The origins of these bacteria in breast milk are still debated, with research exploring various potential pathways, including retrograde backflow and the possibility of a neonatal gut-to-mother mammary gland pathway [
23,
24].
3. Further Steps in the Development of the Gut Microbiota
The gut microbiota of vaginally delivered neonates resembles the vaginal microbiome, with high populations of
Lactobacillus sensu lato and
Prevotella species [
25]; other bacteria, such as the members of the Enterobacteriaceae family, that is
Escherichia or
Klebsiella, and Gram-positive cocci are also present. Occurrence of both last groups, which are facultative pathogens, poses a danger of the neonatal sepsis, especially in low-birth-weight newborns [
11]. As individuals age, the diversity of the gut microbiota expands until it stabilizes into a mature adult composition. This adult microbiota is primarily characterized by dominance of three bacterial phyla and is shaped by factors such as genetics, environment, diet, lifestyle, and gut physiology. By around three years of age, the composition and diversity of a child’s gut microbiota closely resemble those of adults [
26]. As noted before,
Lactobacillus sensu lato species found in the gut microbiota, although in relatively small numbers, appear to be crucial for supporting the overall health of the organism.
4. Lactobacilli in the Adult Gut Microbiota
The human gut microbiome primarily consists of three bacterial phyla: Bacteroidetes (
Porphyromonas, Prevotella), Firmicutes (
Ruminococcus, Clostridium, and
Eubacterium), and Actinobacteria (
Bifidobacterium). While lactobacilli and other facultative anaerobic taxa are present in relatively low quantities within the gut, their regulatory functions in maintaining the gut microbiota are disproportionately significant. This importance has been highlighted by numerous studies focusing on probiotics that contain
Lactobacillus [
27]. Historically one of the mechanisms of the
Lactobacillus—microbiota interactions involve exerting growth-promoting factors for the other microorganisms which was the first idea on probiotics as proposed by Lilly and Stilwell in 1965 [
28].
Lactobacillus sensu lato bacteria, after successful early colonization of the gut, become long-life members of the gut microbiota although their numbers as well numbers of bifidobacteria decrease in elderly persons [
29]. Again, there are multiple clinical trials with
Lactobacillus- (and
Bifidobacterium-) containing probiotics showing that supplementation of the elderly gut microbiota with such bacteria leads to restoration of the gut microbiota composition but may also prevent development of the gut carcinoma [
30]. Besides genetic aberrations and non-coding RNA, disruptions in gut microbiota, known as dysbiosis, play a role in the development of colon cancer. Among various interventions, lactic acid bacteria, including
Lactobacillus sensu lato, have been extensively researched as probiotics for their potential in cancer prevention [
6].
After effectively colonizing the gut, lactobacilli engage in direct communication with human epithelial cells. For instance, they trigger transcriptional modifications in the gut epithelium, leading to changes in the expression of numerous genes within just a few hours following oral administration. Most of the human genes involved in down- or up-regulation by lactobacilli are coding for fatty acid metabolism and cellular growth and development [
31]. In relation to regulation of the gut immune response, lactobacilli induce anti-inflammatory activities related to NF-kappa B complex [
32]. Moreover, animal studies also indicate that presence of
Lactobacillus sensu lato is associated with the growth of beneficial gut microbiota. The phyla Firmicutes and Bacteroidetes form the core microbiota, accounting for over 90% of the known phylogenetic groups present in the healthy gut of breastfed mice, a composition that closely mirrors the core microbiota in human infants [
33]. Lactate produced by microbes strongly induces colonic hyperproliferation in mice, with this effect being most evident after a short-term period of starvation. Additionally, lactate serves as a crucial energy source for small intestinal stem cells, enabling them to maintain their capacity for proliferation and differentiation, as shown in organoid models [
34].
Another significant, although not restricted to this group, function of
Lactobacillus in human gut microbiota which positively influences health is production of vitamin K2 (menaquinone). This property was mentioned already 30 years ago in the fundamental work on its synthesis by Bentley and Meganathan [
35]. Later on, it was revealed that yoghurt-fermenting lactobacilli produce menaquinone-7 [
36]. However, contemporary data, although confirming the important role of the gut bacteria in vitamin K2 synthesis, do not link it to particular genera. This remains to be elucidated in the light of the actual bacterial taxonomy.
Alterations in gut microbiota composition are observed in various disease conditions, including cardiovascular disease, cancer, type 2 diabetes mellitus, obesity, colitis, asthma, psychiatric and inflammatory disorders, gut-brain axis disturbances, and numerous immune-related issues. Consequently, modulating the gut microbiota through probiotic bacteria, particularly those from the
Lactobacillus genus, may help address a variety of health challenges. In fact, a range of degenerative diseases, such as obesity, diabetes, cancer, cardiovascular conditions, liver diseases, and inflammatory bowel disease (IBD), have been shown to be preventable by various
Lactobacillus sensu lato strains from different species [
27].
5. The Role of Lactobacilli in Gut Innate and Local Immunity
Initial interactions with pioneer bacteria may play a crucial role in determining subsequent gut maturation, as well as in shaping metabolic and immune responses, ultimately influencing both short- and long-term health outcomes [
37].
Newborns enter the world with an underdeveloped and naive acquired immune system, and several aspects of their innate mucosal immunity are not fully matured. As a result, the innate immune components found in human milk play a critical role in supporting the mucosal barrier of the infant’s developing gut. Through breastfeeding, mothers supply their infants with numerous protective agents, some of which have been identified as
Lactobacillus sensu lato strains in laboratory infection models [
38].
As with other aspects of the interactions between lactobacilli and the human organism, most of the information on
Lactobacillus sensu lato on immunity comes from studies on probiotic lactobacilli on animal models. There are multiple studies showing that probiotic lactobacilli contribute to the host’s well-being by enhancing the balance of intestinal microflora and potentially strengthening the host’s defense mechanisms. The mechanism by which innate immunity is stimulated in mammals and birds relies on NF-kappaB and p38 MAPK signaling pathways, as demonstrated on mice, cows, pigs and chickens [
39,
40,
41,
42,
43]. It is obvious that this property is not restricted to probiotic strains of lactobacilli, although it is highly individualized among strains.
It is commonly agreed, especially in food science, that intestinal homeostasis in which local gut immunity plays an important role, can be reestablished by consuming supplements that contain fermented dairy products. These products often contain beneficial microbes, such as probiotics, predominantly from the
Lactobacillus sensu lato genera. Multiple studies done on different
Lactobacillus sensu lato strains commonly stress how crucial they are in modulating the gut immune response within both the innate and mucosal immune systems, such as T, B, and NK lymphocytes [
44]. It has been shown that a very broad group of the
Lactobacillus strains representing different species show common characteristics. These include the ability to activate an inflammatory response in macrophages in vitro through mechanisms involving the production of proinflammatory mediators, such as cytokines and reactive oxygen species (ROS), as well as involvement in signaling pathways, including NF-kappaB and TLR2 [
45,
46].
On the other hand, the regulatory functions of lactobacilli in the gut promote the production of anti-inflammatory cytokines, which has also been found [
45] to positively impact chronic gastrointestinal issues. The work of probiotic lactobacilli in modulating the inflammatory response from the gut mucosa has also been demonstrated in patients with inflammatory bowel disease (IBD), highlighting their beneficial role in managing such conditions [
47]. It should be noted that it is commonly accepted that IBD is linked to alterations in the composition and metabolism of the intestinal microbiota, known as dysbiosis, which interact with host genetic mutations and changed immune response to gut microbes. However, a definitive causal link between dysbiosis and IBD has not yet been established in humans [
48].
There are at least two mechanisms by which
Lactobacillus sensu lato may ameliorate chronic gut inflammation. Some
Lactobacillus species possess anti-inflammatory enzymes decomposing ROS, like superoxide dismutase or catalase [
49,
50]. Observations on the gut microbiota composition in IBD patients and especially in pediatric patients with new-onset disease showed its altered composition with reduction of the bacteria belonging to phylum Firmicutes represented also by
Lactobacillus sensu lato, support this idea [
51,
52]. On the other hand, increased populations of Proteobacteria are able to induce gut inflammation [
53,
54]. These data may also indicate an anti-inflammatory role of the lactobacilli in vivo related to their anti-oxidative properties [
50]. There is also another explanation offered for the beneficial role of
Lactobacillus sensu lato probiotics in IBD. Animal studies have shown that the NF-kappaB pathway is inhibited by probiotics, which promotes the activation of immune-modulating regulatory T cells and reduces the presence of proinflammatory effector-memory T cells in the intestinal mucosa [
55].
Immune-related disorders have been connected to a decline in overall bacterial diversity, a reduced presence of
Lactobacillus sensu lato in a healthy gut microbiota, and a rise in potentially harmful bacteria. Additionally, specific bacteria—not including lactobacilli—have been identified as contributors to allergic reactions in both animal studies and human research [
56].
Thus, probiotics, especially those containing
Lactobacillus sensu lato strains, are proposed to offer potential benefits in preventing and treating certain immune-related diseases through the modulation of gut microbiota and the regulation of host mucosal immune function. Nonetheless, their effectiveness has been variable, and additional research is needed to confirm these outcomes [
56,
57].
6. Lactobacilli in the Vaginal Microbiota
Undoubtedly, the most completely documented leading role of the
Lactobacillus sensu lato bacteria in keeping homeostasis and protecting healthy status occurs in human vagina where lactobacilli act solely as both pioneering and sentinel organisms forming and keeping normal vaginal microbiota composition in the majority of women of childbearing age [
58]. Vaginal lactobacilli are a crucial defense against invading pathogens by producing bactericidal compounds and metabolites, as well as by modulating the innate immune responses in the vagina [
59]. There are suggestions that not all
Lactobacillus species residing in the vagina are equal in their protective role and that
L. crispatus is the most active [
60] in this respect. Conversely, a very recent study by Hassan et al. indicates that reduced species diversity, combined with an increased abundance of
L. iners, L. gasseri and
G. vaginalis, could be linked to female infertility [
61].
The substantial need to discriminate among the species
Lactobacillus in the vagina and roles of the new taxa and their taxonomic positions should be re-evaluated and in the light of the extensive studies on vaginal microbiota presented by Ravel
et al., that there are at least several types of the microbiota with different predominant species of the
Lactobacillus sensu lato [
62]. The study by Ravel et al. documented that not all species of the
Lactobacillus genera possess ability to colonize the vaginal epithelium, and
L. crispatus, L. iners, L. gasseri and
L. jensenii are listed as the most typical members of this community, however with different influences in vaginal health, as shown above. Still, there are very interesting differences in composition of the
Lactobacillus populations related to a mix of cultural, behavioral, genetic and other as-yet-unknown factors [
62]. Most probably these differences are caused by unknown variants in specific receptors in vaginal epithelium as well as by various foods since gut microbiota serve as a reservoir of the vaginal lactobacilli. In fact, the human vaginal microbiota is typically dominated by one or two of the five primary lactobacilli species and is distinguished by a pH level below 4.5 [
63].
High populations of lactobacilli are constantly supplemented and supported by these strains which pass from GI tract over perianal, perineal and vulvar skin to the vaginal orifice thanks to unique mechanisms driven by specific genes [
64,
65]. It is noteworthy that these mechanisms have been utilized in constructing and implementing vaginal probiotic products for the oral delivery [
66,
67,
68]. The abundance of the vaginal lactobacilli is also regulated by hormonal interplay since it increases in puberty and further during pregnancy [
69] and decreases during menopause. Most probably other factors are also involved in acquisition of the lactobacilli since it appeared recently that they appear before menarche. It was shown that the microbiota of most girls before menarche was predominantly composed of various
Lactobacillus species, observed during the early to mid-puberty stages [
70].
7. Innate Immunity in the Vagina
Many studies continue to indicate that a vaginal bacterial community dominated by lactobacilli plays a significant role in driving the innate immune response [
71]. The health of both pregnant and non-pregnant women is supported by a higher concentration of vaginal lactobacilli. Their resistance to infections is also enhanced under such conditions [
72].
When established in the vaginal epithelium, lactobacilli successfully inhibit other bacteria using several well-known factors such as lowering pH, thanks to production of a variety of the organic acids, bacteriocin production and dissolving biofilm structures [
73,
74]. Low pH of the vaginal secretion is linked to the lactic acid produced by all
Lactobacillus species, but, in fact, except for lactic acid, there are multiple organic acids produced by lactobacilli in the vagina at lower amounts but possessing much stronger antimicrobial activities [
75,
76]. It is generally accepted that lactobacilli produce lactic acid from vaginal glycogen, but this theory is not supported with high quality studies. Remarkably, despite more than a hundred years since the strong association between glycogen, lactobacilli, and lactic acid in the vagina was established, we still have limited knowledge about how lactobacilli metabolize glycogen [
77]. Even with the intensive sequencing studies, molecular mechanisms of
Lactobacillus predominance in vaginal microbiota remain unexplored.
Other elements that play a role in vaginal defense are mannose-binding lectin (MBL), vaginal antimicrobial peptides (AMPs), and immunoglobulins A and G (IgA and IgG).
The presence of protective
Lactobacillus species in the vagina leads to reduced activation of vaginal epithelial cells. Numerous epidemiological studies have shown that women with a vaginal microbiota predominantly consisting of
L. crispatus have lower levels of pro-inflammatory cytokines, such as interleukin (IL)-1α and IL-1β, as well as chemokines like IL-8, interferon-gamma inducible protein (IP-10), and macrophage inflammatory protein (MIP)-3α in their cervicovaginal secretions [
78].
8. Adaptive Immunity
The female genital tract contains most elements of both the adaptive and innate mucosal immune systems. Unlike other mucosal areas, the vagina has a limited amount of local lymphoid tissue. While other mucosal sites mainly produce IgA and IgM, the predominant immunoglobulin secreted in the vagina and cervix is IgG [
79]. Vaginal epithelial cells generate various antimicrobial substances and are equipped with membrane-bound Toll-like receptors that detect pathogen-associated molecular patterns. This detection triggers the production of pro-inflammatory cytokines and antigen-specific immune responses. Additionally, the endocervix and vagina can locally produce IgG and IgA antibodies in response to infections [
69,
80].
We need to agree with Vaneechoutte [
63] that, despite over a century of research, significant debates persist on fundamental issues, with ongoing publication of controversial findings. Given the substantial effects of vaginal microbiology on women’s health and pregnancy outcomes, it is crucial to address these controversies. Doing so will enhance our understanding of vaginal microbiology and its relationship with vaginal immunity, leading to improvements in vaginal probiotics and approaches aimed at helping to maintain or restore
Lactobacillus dominance.
9. Conclusions
The name Lactobacillus is broadly recognized and utilized by the medical community, the food and health-related industries, and the general public, and is also featured in national and international regulations. Despite being a traditional term that encompasses multiple genera and numerous species, it remains widely accepted. The bacteria hindered efforts to study the ecology, physiology, evolution, and practical uses of this significant group of organisms. In reality, these bacteria are genetically distinct and exhibit considerable diversity in their metabolism, ecology, and functions. As a result, they can no longer be grouped within the same genus and thus a new taxonomy has been proposed for them, and its adoption will stimulate a new wave of research on relations between taxonomic position and characteristics of the bacterial strains formerly placed in Lactobacillus genus. This will undoubtedly yield new insights into the significance of these bacteria for our health, even though many of them will be assigned new names.
Author Contributions
P.B.H. conceptualized and designed the study and data collection forms, performed the initial searches, extracted the data, conducted the literature review on lactobacilli, drafted the initial manuscript, and approved the final manuscript as submitted; M.G and W.P. conducted the literature review, drafted, and edited the manuscript; M.S. conceptualized and designed the study, reviewed and revised the manuscript, and approved the final manuscript as submitted.
Funding
This work was supported by the National Centre for Research and Development, grant no. RPMP.01.02.01-12-0413/17.
Acknowledgments
The authors wish to thank Doctor Piotr Kochan for his help in correcting the style of this manuscript.
Conflicts of Interest
The authors declare no conflict of interest.
References
- Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; Watanabe, K.; Wuyts, S.; Felis, G.E.; Gänzle, M.G.; Lebeer, S. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef] [PubMed]
- O’Callaghan, J.; O’Toole, P.W. Lactobacillus: host-microbe relationships. Curr. Top Microbiol. Immunol. 2013, 358, 119–5. [Google Scholar] [PubMed]
- De Angelis, M.; Calasso, M.; Cavallo, N.; Di Cagno, R.; Gobbetti, M. Functional proteomics within the genus Lactobacillus. Proteomics 2016, 16, 946–62. [Google Scholar] [CrossRef] [PubMed]
- Seddik, H.A.; Bendali, F.; Gancel, F.; Fliss, I.; Spano, G.; Drider, D. Lactobacillus plantarum and Its Probiotic and Food Potentialities. Probiotics Antimicrob. Proteins 2017, 9, 111–122. [Google Scholar] [CrossRef]
- Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef] [PubMed]
- Cavaillon, J.M.; Legout, S. Centenary of the death of Elie Metchnikoff: a visionary and an outstanding team leader. Microbes Infect. 2016, 18, 577–594. [Google Scholar] [CrossRef]
- Hornef, M.W.; Torow, N. ‘Layered immunity’ and the ‘neonatal window of opportunity’—timed succession of non-redundant phases to establish mucosal host-microbial homeostasis after birth. Immunology 2020, 159, 15–25. [Google Scholar] [CrossRef]
- Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M. Intestinal permeability—a new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef]
- Strus, M.; Helwich, E.; Lauterbach, R.; Rzepecka-Węglarz, B.; Nowicka, K.; Wilińska, M.; Szczapa, J.; Rudnicka, M.; Sławska, H.; Szczepański, M.; Waśko, A.; Mikołajczyk-Cichońska, A.; Tomusiak-Plebanek, A.; Heczko, P.B. Effects of oral probiotic supplementation on gut Lactobacillus and Bifidobacterium populations and the clinical status of low-birth-weight preterm neonates: a multicenter randomized, double-blind, placebo-controlled trial. Infect Drug Resist. 2018, 11, 1557–1571. [Google Scholar] [CrossRef]
- Golińska, E.; Strus, M.; Tomusiak-Plebanek, A.; Więcek, G.; Kozień, Ł.; Lauterbach, R.; Pawlik, D.; Rzepecka-Węglarz, B.; Kędzierska, J.; Dorycka, M.; Heczko, P.B. Coagulase-Negative Staphylococci Contained in Gut Microbiota as a Primary Source of Sepsis in Low- and Very Low Birth Weight Neonates. J. Clin. Med. 2020, 9, 2517. [Google Scholar] [CrossRef] [PubMed]
- Wójkowska-Mach, J.; Chmielarczyk, A.; Strus, M.; Lauterbach, R.; Heczko, P. Neonate Bloodstream Infections in Organization for Economic Cooperation and Development Countries: An Update on Epidemiology and Prevention. J. Clin. Med. 2019, 8, 1750. [Google Scholar] [CrossRef] [PubMed]
- Urushiyama, D.; Suda, W.; Ohnishi, E.; Araki, R.; Kiyoshima, C.; Kurakazu, M.; Sanui, A.; Yotsumoto, F.; Murata, M.; Nabeshima, K.; Yasunaga, S.; Saito, S.; Nomiyama, M.; Hattori, M.; Miyamoto, S.; Hata, K. Microbiome profile of the amniotic fluid as a predictive biomarker of perinatal outcome. Sci. Rep. 2017, 7, 12171. [Google Scholar] [CrossRef] [PubMed]
- Satokari, R.; Grönroos, T.; Laitinen, K.; Salminen, S.; Isolauri, E. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett. Appl. Microbiol. 2009, 48, 8–12. [Google Scholar] [CrossRef] [PubMed]
- Rehbinder, E.M.; Lødrup Carlsen, K.C.; Staff, A.C.; Angell, I.L.; Landrø, L.; Hilde, K.; Gaustad, P.; Rudi, K. Is amniotic fluid of women with uncomplicated term pregnancies free of bacteria? Am. J. Obstet. Gynecol. 2018, 219, 289–e1. [Google Scholar] [CrossRef]
- Liu, Y.; Li, X.; Zhu, B.; Zhao, H.; Ai, Q.; Tong, Y.; Qin, S.; Feng, Y.; Wang, Y.; Wang, S.; Ma, J.; Yang, H. Midtrimester amniotic fluid from healthy pregnancies has no microorganisms using multiple methods of microbiologic inquiry. Am. J. Obstet. Gynecol. 2020, 223, 248–e1. [Google Scholar] [CrossRef] [PubMed]
- Bagg, J.; MacFarlane, T.W.; Poxton, I.R.; Smith, A.J. Essentials of microbiology for dental students. 2006 (No. Ed. 2). Oxford university press.
- Hegde, S.; Munshi, A.K. Influence of the maternal vaginal microbiota on the oral microbiota of the newborn. J. Clin. Pediatr. Dent. 1998, 22, 317–21. [Google Scholar] [PubMed]
- Mändar, R.; Mikelsaar, M. Transmission of mother’s microflora to the newborn at birth. Biol Neonate. 1996, 69, 30–5. [Google Scholar] [CrossRef] [PubMed]
- He, Q.; Kwok, L.Y.; Xi, X.; Zhong, Z.; Ma, T.; Xu, H.; Meng, H.; Zhao, F.; Zhang, H. The meconium microbiota shares more features with the amniotic fluid microbiota than the maternal fecal and vaginal microbiota. Gut Microbes 2020, 12, 1794266. [Google Scholar] [CrossRef]
- Baäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef]
- Soto, A.; Martín, V.; Jiménez, E.; Mader, I.; Rodríguez, J.M.; Fernández, L. Lactobacilli and bifidobacteria in human breast milk: influence of antibiotherapy and other host and clinical factors. J. Pediatr. Gastroenterol. Nutr. 2014, 59, 78–88. [Google Scholar] [CrossRef]
- Rodríguez, J.M. The origin of human milk bacteria: is there a bacterial entero-mammary pathway during late pregnancy and lactation? Adv. Nutr. 2014, 5, 779–84. [Google Scholar] [CrossRef] [PubMed]
- Lyons, K.E.; Ryan, C.A.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020, 12, 1039. [Google Scholar] [CrossRef]
- Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–5. [Google Scholar] [CrossRef]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed]
- Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. Biomed Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef]
- Gupta, V.; Garg, R. Probiotics. Indian J. Med. Microbiol. 2009, 27, 202–9. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Jazwinski, S.M. The Gut Microbiota and Healthy Aging: A Mini-Review. Gerontology 2018, 64, 513–520. [Google Scholar] [CrossRef]
- Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef]
- Troost, F.J.; van Baarlen, P.; Lindsey, P.; Kodde, A.; de Vos, W.M.; Kleerebezem, M.; Brummer, R.J. Identification of the transcriptional response of human intestinal mucosa to Lactobacillus plantarum WCFS1 in vivo. BMC Genomics 2008, 9, 374. [Google Scholar] [CrossRef]
- van Baarlen, P.; Troost, F.J.; van Hemert, S.; van der Meer, C.; de Vos, W.M.; de Groot, P.J.; Hooiveld, G.J.; Brummer, R.J.; Kleerebezem, M. Differential NF-kappaB pathways induction by Lactobacillus plantarum in the duodenum of healthy humans correlating with immune tolerance. Proc. Natl. Acad. Sci. USA 2009, 106, 2371–6. [Google Scholar] [CrossRef]
- Liu, Y.; Tian, X.; He, B.; Hoang, T.K.; Taylor, C.M.; Blanchard, E.; Freeborn, J.; Park, S.; Luo, M.; Couturier, J.; Tran, D.Q.; Roos, S.; Wu, G.; Rhoads, J.M. Lactobacillus reuteri DSM 17938 feeding of healthy newborn mice regulates immune responses while modulating gut microbiota and boosting beneficial metabolites. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G824–G838. [Google Scholar] [CrossRef] [PubMed]
- Allaire, J.M.; Crowley, S.M.; Law, H.T.; Chang, S.Y.; Ko, H.J.; Vallance, B.A. The Intestinal Epithelium: Central Coordinator of Mucosal Immunity. Trends Immunol. 2018, 39, 677–696. [Google Scholar] [CrossRef]
- Bentley, R.; Meganathan, R. Biosynthesis of vitamin K (menaquinone) in bacteria. Microbiol. Rev. 1982, 46, 241–80. [Google Scholar] [CrossRef] [PubMed]
- Plaza, S.M. The anticancer effects of vitamin K. Altern. Med. Rev. 2003, 8, 303–318. [Google Scholar]
- Rook, G.A.; Lowry, C.A.; Raison, C.L. Hygiene and other early childhood influ- ences on the subsequent function of the immune system. Brain Res. ( 2014.
- Newburg, D.S. Innate immunity and human milk. J. Nutr. 2005, 135, 1308–12. [Google Scholar] [CrossRef]
- Cheng, C.; Zhang, L.; Mu, J.; Tian, Q.; Liu, Y.; Ma, X.; Fu, Y.; Liu, Z.; Li, Z. Effect of Lactobacillus johnsonii Strain SQ0048 on the TLRs-MyD88/NF-κB Signaling Pathway in Bovine Vaginal Epithelial Cells. Front. Vet Sci. 2021, 8, 670949. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Lü, X.; Man, C.; Han, L.; Shan, Y.; Qu, X.; Liu, Y.; Yang, S.; Xue, Y.; Zhang, Y. Lactobacillus acidophilus induces cytokine and chemokine production via NF-κB and p38 mitogen-activated protein kinase signaling pathways in intestinal epithelial cells. Clin. Vaccine Immunol. 2012, 19, 603–8. [Google Scholar] [CrossRef]
- Kim, Y.G.; Ohta, T.; Takahashi, T.; Kushiro, A.; Nomoto, K.; Yokokura, T.; Okada, N.; Danbara, H. Probiotic Lactobacillus casei activates innate immunity via NF-kappaB and p38 MAP kinase signaling pathways. Microbes Infect. 2006, 8, 994–1005. [Google Scholar] [CrossRef]
- Shin, D.; Chang, S.Y.; Bogere, P.; Won, K.; Choi, J.Y.; Choi, Y.J.; Lee, H.K.; Hur, J.; Park, B.Y.; Kim, Y.; Heo, J. Beneficial roles of probiotics on the modulation of gut microbiota and immune response in pigs. PLoS One 2019, 14, e0220843. [Google Scholar] [CrossRef] [PubMed]
- Richards, P.J.; Flaujac Lafontaine, G.M.; Connerton, P.L.; Liang, L.; Asiani, K.; Fish, N.M.; Connerton, I.F. Galacto-Oligosaccharides Modulate the Juvenile Gut Microbiome and Innate Immunity To Improve Broiler Chicken Performance. mSystems 2020, 5, e00827–19. [Google Scholar] [CrossRef] [PubMed]
- Forsythe, P.; Bienenstock, J. Immunomodulation by commensal and probiotic bacteria. Immunol Invest. 2010, 39, 429–48. [Google Scholar] [CrossRef]
- Rocha-Ramírez, L.M.; Pérez-Solano, R.A.; Castañón-Alonso, S.L.; Moreno Guerrero, S.S.; Ramírez Pacheco, A.; García Garibay, M.; Eslava, C. Probiotic Lactobacillus Strains Stimulate the Inflammatory Response and Activate Human Macrophages. J. Immunol. Res. 2017, 2017, 4607491. [Google Scholar] [CrossRef] [PubMed]
- Marcinkiewicz, J.; Ciszek, M.; Bobek, M.; Strus, M.; Heczko, P.B.; Kurnyta, M.; Biedroń, R.; Chmielarczyk, A. Differential inflammatory mediator response in vitro from murine macrophages to lactobacilli and pathogenic intestinal bacteria. Int. J. Exp. Pathol. 2007, 88, 155–64. [Google Scholar] [CrossRef] [PubMed]
- Jakubczyk, D.; Leszczyńska, K.; Górska, S. The Effectiveness of Probiotics in the Treatment of Inflammatory Bowel Disease (IBD)-A Critical Review. Nutrients 2020, 12, 1973. [Google Scholar] [CrossRef]
- Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut microbiota and IBD: causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef] [PubMed]
- Strus, M.; Brzychczy-Włoch, M.; Kochan, P.; Heczko, P. Nadtlenek wodoru, wytwarzany przez bakterie z rodzaju Lactobacillus, jako czynnik regulujacy mikroflore pochwy [Hydrogen peroxide produced by Lactobacillus species as a regulatory molecule for vaginal microflora]. Med. Dosw Mikrobiol. 2004, 56, 67–77. [Google Scholar] [PubMed]
- Tomusiak-Plebanek, A.; Heczko, P.; Skowron, B.; Baranowska, A.; Okoń, K.; Thor, P.J.; Strus, M. Lactobacilli with superoxide dismutase-like or catalase activity are more effective in alleviating inflammation in an inflammatory bowel disease mouse model. Drug Des. Devel. Ther. 2018, 12, 3221–3233. [Google Scholar] [CrossRef]
- Gevers, D.; Kugathasan, S.; Denson, L.A.; Vázquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; Morgan, X.C.; Kostic, A.D.; Luo, C.; González, A.; McDonald, D.; Haberman, Y.; Walters, T.; Baker, S.; Rosh, J.; Stephens, M.; Heyman, M.; Markowitz, J.; Baldassano, R.; Griffiths, A.; Sylvester, F.; Mack, D.; Kim, S.; Crandall, W.; Hyams, J.; Huttenhower, C.; Knight, R.; Xavier, R.J. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef] [PubMed]
- Fyderek, K.; Strus, M.; Kowalska-Duplaga, K.; Gosiewski, T.; Wedrychowicz, A.; Jedynak-Wasowicz, U.; Sładek, M.; Pieczarkowski, S.; Adamski, P.; Kochan, P.; Heczko, P.B. Mucosal bacterial microflora and mucus layer thickness in adolescents with inflammatory bowel disease. World J. Gastroenterol. 2009, 15, 5287–94. [Google Scholar] [CrossRef] [PubMed]
- Pilarczyk-Zurek, M.; Chmielarczyk, A.; Gosiewski, T.; Tomusiak, A.; Adamski, P.; Zwolinska-Wcislo, M.; Mach, T.; Heczko, P.B.; Strus, M. Possible role of Escherichia coli in propagation and perpetuation of chronic inflammation in ulcerative colitis. BMC Gastroenterol. 2013, 13, 61. [Google Scholar] [CrossRef] [PubMed]
- Pilarczyk-Zurek, M.; Strus, M.; Adamski, P.; Heczko, P.B. The dual role of Escherichia coli in the course of ulcerative colitis. BMC Gastroenterol. 2016, 16, 128. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Fatheree, N.Y.; Mangalat, N.; Rhoads, J.M. Lactobacillus reuteri strains reduce incidence and severity of experimental necrotizing enterocolitis via modulation of TLR4 and NF-κB signaling in the intestine. Am J Physiol Gastrointest Liver Physiol. 2012, 302, G608–17. [Google Scholar] [CrossRef] [PubMed]
- Munyaka, P.M.; Khafipour, E.; Ghia, J.E. External influence of early childhood establishment of gut microbiota and subsequent health implications. Front Pediatr. 2014, 2, 109. [Google Scholar] [CrossRef]
- Ashraf, R.; Shah, N.P. Immune system stimulation by probiotic microorganisms. Crit Rev Food Sci Nutr. 2014, 54, 938–56. [Google Scholar] [CrossRef]
- Amabebe, E.; Anumba, D.O.C. The Vaginal Microenvironment: The Physiologic Role of Lactobacilli. Front Med (Lausanne). 2018, 5, 181. [Google Scholar] [CrossRef]
- Onderdonk, A.B.; Delaney, M.L.; Fichorova, R.N. The Human Microbiome during Bacterial Vaginosis. Clin Microbiol Rev 2016, 29, 223–38. [Google Scholar] [CrossRef] [PubMed]
- Muzny, C.A.; Blanchard, E.; Taylor, C.M.; Aaron, K.J.; Talluri, R.; Griswold, M.E.; Redden, D.T.; Luo, M.; Welsh, D.A.; Van Der Pol, W.J.; Lefkowitz, E.J.; Martin, D.H.; Schwebke, J.R. Identification of Key Bacteria Involved in the Induction of Incident Bacterial Vaginosis: A Prospective Study. J Infect Dis. 2018, 218, 966–978. [Google Scholar] [CrossRef]
- Hasan, Z.; Netherland, M.; Hasan, N.A.; Begum, N.; Yasmin, M.; Ahmed, S. 2024.1390088. eCollection 2024. An insight into the vaginal microbiome of infertile women in Bangladesh using metagenomic approach. Front Cell Infect Microbiol. 2024, 14, 1390088. [Google Scholar] [CrossRef]
- Ravel, J.; Gajer, P.; Abdo, Z.; Schneider, G.M.; Koenig, S.S.K.; McCulle, S.L.; et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci USA 2011, 108, 4680–7. [Google Scholar] [CrossRef] [PubMed]
- Vaneechoutte, M. The human vaginal microbial community. Res Microbiol. 2017, 168, 811–825. [Google Scholar] [CrossRef] [PubMed]
- El Aila, N.A.; Tency, I.; Claeys, G.; Verstraelen, H.; Saerens, B.; Santiago, G.L.; De Backer, E.; Cools, P.; Temmerman, M.; Verhelst, R.; Vaneechoutte, M. Identification and genotyping of bacteria from paired vaginal and rectal samples from pregnant women indicates similarity between vaginal and rectal microflora. BMC Infect Dis. 2009, 9, 167. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Gustafsson, R.J.; Ahrné, S.; Jeppsson, B.; Benoni, C.; Olsson, C.; Stjernquist, M.; Ohlsson, B. The Lactobacillus flora in vagina and rectum of fertile and postmenopausal healthy Swedish women. BMC Womens Health. 2011, 11, 17. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Reid, G.; Charbonneau, D.; Erb, J.; Kochanowski, B.; Beuerman, D.; Poehner, R.; Bruce, A.W. Oral use of Lactobacillus rhamnosus GR-1 and L. fermentum RC-14 significantly alters vaginal flora: randomized, placebo-controlled trial in 64 healthy women. FEMS Immunol Med Microbiol. 2003, 35, 131–4. [Google Scholar] [CrossRef]
- Heczko, P.B.; Tomusiak, A.; Adamski, P.; Jakimiuk, A.J.; Stefański, G.; Mikołajczyk-Cichońska, A.; Suda-Szczurek, M.; Strus, M. Supplementation of standard antibiotic therapy with oral probiotics for bacterial vaginosis and aerobic vaginitis: a randomised, double-blind, placebo-controlled trial. BMC Womens Health. 2015, 15, 115. [Google Scholar] [CrossRef]
- Koirala, R.; Gargari, G.; Arioli, S.; Taverniti, V.; Fiore, W.; Grossi, E.; Anelli, G.M.; Cetin, I.; Guglielmetti, S. Effect of oral consumption of capsules containing Lactobacillus paracasei LPC-S01 on the vaginal microbiota of healthy adult women: a randomized, placebo-controlled, double-blind crossover study. FEMS Microbiol Ecol. 2020, 96, fiaa084. [Google Scholar] [CrossRef]
- Witkin, S.S.; Linhares, I.M. Why do lactobacilli dominate the human vaginal microbiota? BJOG. 2017, 124, 606–611. [Google Scholar] [CrossRef]
- Hickey, R.J.; Zhou, X.; Settles, M.L.; Erb, J.; Malone, K.; Hansmann, M.A.; Shew, M.L.; Van Der Pol, B.; Fortenberry, J.D.; Forney, L.J. Vaginal microbiota of adolescent girls prior to the onset of menarche resemble those of reproductive-age women. mBio. 2015, 6, e00097–15. [Google Scholar] [CrossRef] [PubMed]
- Smith, S.B.; Ravel, J. The vaginal microbiota, host defence and reproductive physiology. J Physiol. 2017, 595, 451–463. [Google Scholar] [CrossRef]
- Witkin, S.S.; Linhares, I.M.; Giraldo, P. Bacterial flora of the female genital tract: function and immune regulation. Best Pract Res Clin Obstet Gynaecol. 2007, 21, 347–54. [Google Scholar] [CrossRef] [PubMed]
- Saunders, S.; Bocking, A.; Challis, J.; Reid, G. Effect of Lactobacillus challenge on Gardnerella vaginalis biofilms. Colloids Surf B Biointerfaces. 2007, 55, 138–42. [Google Scholar] [CrossRef] [PubMed]
- Qian, Z.; Zhu, H.; Zhao, D.; Yang, P.; Gao, F.; Lu, C.; Yin, Y.; Kan, S.; Chen, D. Probiotic Lactobacillus sp. Strains Inhibit Growth, Adhesion, Biofilm Formation, and Gene Expression of Bacterial Vaginosis-Inducing Gardnerella vaginalis. Microorganisms. 2021, 9, 728. [Google Scholar] [CrossRef] [PubMed]
- Neal-McKinney, J.M.; Lu, X.; Duong, T.; Larson, C.L.; Call, D.R.; Shah, D.H.; Konkel, M.E. Production of organic acids by probiotic lactobacilli can be used to reduce pathogen load in poultry. PLoS One. 2012, 7, e43928. [Google Scholar] [CrossRef]
- Bukhari, S.A.; Salman, M.; Numan, M.; Javed, M.R.; Zubair, M.; Mustafa, G. Characterization of antifungal metabolites produced by Lactobacillus plantarum and Lactobacillus coryniformis isolated from rice rinsed water. Mol Biol Rep. 2020, 47, 1871–1881. [Google Scholar] [CrossRef] [PubMed]
- Nunn, K.L.; Forney, L.J. Unraveling the dynamics of the human vaginal microbiome. Yale J Biol Med 2016, 89, 331e7. [Google Scholar]
- Masson, L.; Barnabas, S.; Deese, J.; et al. Inflammatory cytokine biomarkers of asymptomatic sexually transmitted infections and vaginal dysbiosis: a multicentre validation study. Sex Transm Infect 2019, 95, 5–12. [Google Scholar] [CrossRef]
- Naz, R.K. Female genital tract immunity: distinct immunological challenges for vaccine development. J Reprod Immunol. 2012, 93, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Linhares, I.M.; Giraldo, P.C.; Baracat, E.C. New findings about vaginal bacterial flora. Rev Assoc Med Bras (1992). 2010, 56, 370–4. [Google Scholar] [CrossRef] [PubMed]
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).