Next Article in Journal
Prevalence of mecA, mecC and Panton-Valentine-Leukocidin Genes in Clinical Isolates of Coagulase Positive Staphylococci from Dermatological Canine Patients
Next Article in Special Issue
Role of Probiotics and Diet in the Management of Neurological Diseases and Mood States: A Review
Previous Article in Journal
Endocrine Disrupting Compounds (Nonylphenol and Bisphenol A)–Sources, Harmfulness and Laccase-Assisted Degradation in the Aquatic Environment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Role of Genital Tract Bacteria in Promoting Endometrial Health in Cattle

1
Department of Biomedicine, Institute of Veterinary Sciences, University of Tiaret, Tiaret 14000, Algeria
2
Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre 00265, Saint Kitts and Nevis
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(11), 2238; https://doi.org/10.3390/microorganisms10112238
Submission received: 12 October 2022 / Revised: 8 November 2022 / Accepted: 10 November 2022 / Published: 12 November 2022
(This article belongs to the Special Issue Beneficial Microbes: Food, Mood and Beyond)

Abstract

:
Microbiota regulate endometrial health in cattle. It is important to know what a ‘good’ microbiome is, in order to understand pathogeneses of uterine disease. Given that microbial influx into the genital tract of cows at calving is unavoidable, exploring the involvement of genital tract bacteria in promoting endometrial health is warranted. The dysbiosis of endometrial microbiota is associated with benign and malign uterine diseases. The present review discusses current knowledge about the altered endometrial microbiome and the implications of this modulation on endometrial inflammation, ovarian activity, fecundation, pregnancy, and postpartum complications. Intravaginal administration of symbiotic microbes in cattle is a realistic alternative to antibiotic and hormone therapy to treat uterine disease. Genital microbial diversity can be modeled by nutrition, as the energy balance would improve the growth of specific microbial populations. It may be that probiotics that alter the endometrial microbiome could provide viable alternatives to existing therapies for uterine disease in cattle.

1. Introduction

Mammalian organs and systems harbor a complex yet dynamic microbiome, especially genital and digestive tracts. In cattle, the genital tract microbiome exerts a profound influence on endometrial health, homeostasis, and fertility. Many factors contribute to the colonization of the uterus by a diverse microbiome. The gut is considered as one of the main drivers of bacteria, viruses, and protozoa that colonize the genital tract [1]. Gut microbiota provide essential volatile fatty acids and amino acids that were implicated in sexual communication in mice [2,3]. Rumen microbiota produce enzymes to break down complex feed molecules into simpler ones assimilable in the intestines. In turn, the host provides optimal condition for growth and proliferation of the microbes [4]. We believe that a similar relationship exists in the genital tract of cattle. Biomolecules such as vaginal mucus or host defense proteins play a crucial role in modulating immune responses to these microorganisms. The genital tract microbiota offer many benefits to the host, through a range of physiological functions such as maintaining endometrial epithelial integrity, protecting against pathogens, and regulating host immunity [5,6,7]. For instance, in women, Lactobacillus abundant in the vagina protect the vaginal mucosa by preventing pathogen adhesion through a competition effect [7,8]. In this symbiotic relationship, Lactobacillus utilize genital tract secretions such as mucin carbohydrates as a source of nutrition. In turn, Lactobacillus secrete immune active molecules such as lactic acid and hydrogen peroxide that prevent pathogenic bacterial proliferation [9,10]. Recent advances in microbiology now allow identification of all bacteria present in tissues or organs to the level of phyla or genus. Gene analysis based on the well-conserved 16S ribosomal ribonucleic acid (rRNA) region allowed to accurately detect microbes in organs such as the pregnant uterus previously thought sterile. Bacteria were detected in the uterus of virgin heifers and pregnant cows that were clinically healthy with no risk or gestational complications [11]. Vaginal and uterine microbiome load and diversity are highly dynamic and are influenced by factors such as origin of contamination, delivery mode, nutrition, and postpartum complications [12]. Fecal matter contamination and vaginal microbiomes are the main sources for endometrial colonization contributing to the normal genital microbiome of cows and possible infections [1,5,13]. The sequencing of the V3–V4 region of the 16S rRNA of ectocervicovaginal lavage revealed that the normal vaginal microbiome comprises bacterial phyla Bacteroidetes, Fusobacteria, and Proteobacteria, in association with Archaeal order Desulfurococcales [14,15]. Lactobacillus spp. are present at low rates in 90% of cows postpartum [14,16]. The natural microbiome is beneficial to the host by producing a biofilm, in association with the host secretions such as vaginal mucus that protect the underlying tissue from pathogen invasion [14]. Lactobacillus can co-aggregate with pathogens, which prevent them from adhering to their receptors or ligands in the host’s mucosa. In addition, some of these bacteria produce lactic acid, which reduces vaginal pH and thereby interferes with pathogen proliferation and survival [17]. At normal concentration, lactic acid can inactivate, in vitro, different pathogens, including Chlamydia trachomatis, Neisseria gonorrhoeae, and Escherichia coli (E.coli) [18,19,20]. Changes of the natural balance within populations of the genital microbiome led to dysbiosis and genital diseases which compromise female’s fertility [15,21,22,23,24]. For instance, Gardnerella vaginalis, highly pathogenic bacteria, produce extracellular sialidase, a powerful enzyme that hydrolyzes sialic acid from sialoglycoproteins in bioactive molecules such as secreted Immunoglobulin A (IgA), therefore interfering with the efficiency of the immune system and increasing the risk of infection with other pathogens [25]. The presence of Chlamydia trachomatis, Gardnerella vaginalis, Ureaplasma species, and Gram-negative stains in women’s genital tract would disrupt fertility [24]. Studies report that many factors affect microbial load and diversity; however, few are those describing how the microbiome affects animal fertility. It is known that pheromones which play a vital role in mating are produced, in part, by the vaginal microbiome of different species, including cows [26,27]. In the present paper we review the interaction of genital microbiomes with the host tissues. We aim to explore how cattle fertility is affected by the genital microbiome and suggest possible strategies to improve reproduction and health management, including the use of prebiotics and probiotics. The present paper is not a systematic review; however, search and selection of implicated references were based on relevance to the topic and prioritizing recent published papers.

2. Genital Microbiome and Modulation of Uterine Inflammation

During calving, dilatation of the cervix results in bacterial influx and contamination of the uterus. For cows that develop endometritis, the bacteria present in the uterus postpartum may either be the etiological agents or secondary infections [28]. Molecular-based methods targeting the 16S rRNA gene in vaginal and uterine samples revealed that the high prevalence of pathogenic bacteria such as Fusobacterium and Corynebacterium was commonly associated with metritis, endometritis, and infertility [29,30]. However, the presence of other bacterial species is crucial in the regulation of endometrial inflammation. For instance, Lactobacillus interferes with the secretion of proinflammatory cytokines stimulated by E. coli [31]. In vivo, combining Lactobacillus and Pediococcus cultures resulted in robust control of an endometrial inflammatory response by E. coli [32]. In addition, Lactobacillus sp produce bioactive molecules such as lactic acid and hydrogen peroxide, which inhibit growth of Staphylococcus aureus (S. aureus) and Trueperella pyogenes (T. pyogenes) that are commonly isolated from cattle suffering from uterine disease [33,34]. Lactic acid is a powerful acidic substance that can reach into sensitive microbes without specific receptors and increase cytosol acidity, leading to bacterial death [35]. Thus, a strategy of using Lactobacillus as a probiotic peripartum would likely reduce postpartum uterine disease and improve fertility.

3. Genital Microbiome and Cyclicity

It is well documented that endocrine hormones controlling the estrous cyclicity of cows influence the genital tract microbiome as well as its diversity [36]. In turn, genital microbiota modulates reproductive cycle hormonal profiles [37]. During the follicular phase, high estradiol concentrations lower the pH of endometrial secretions [38]. Thus, estradiol alters microbial diversity in the vagina during the follicular phase [39]. Therefore, it seems that the microbiome present during a specific time influences the estrous cyclicity and quality of developing oocytes [2,37]. Similarly, when present Lactobacillus decreases the vaginal pH and promotes reproductive function by suppressing infectious pathogens, improving oocyte quality and promoting luteal function [2,40,41]. During disease, if the uterus is infected by lipopolysaccharide (LPS)-producing bacteria, such as E. coli, an important inflammatory reaction is triggered in the endometrial tissue and LPS is accumulated in the antrum [42,43]. Furthermore, granulosa cells have specific receptors for LPS, Toll-like receptors 4 (TLR-4), which recognize and respond to LPS, thus triggering an inflammatory reaction in the follicular cells which compromise steroidogenic activity and the development of oocytes through the inhibition of mitotic activity [44,45]. Clinical studies have confirmed that severe uterine contamination was associated with smaller follicles and corpora lutea resulting in low peripheral plasma concentrations of estradiol and progesterone [41,46]. This would likely result in subfertility in cattle.

4. Genital Microbiome and Breeding Management

The genital tract microbiome present when oocytes develop is thought to also influence normal sperm functions and fertilization capabilities, thereby influencing conception [2]. Like bacteria, sperm are deemed foreign bodies by the genital tract immune system and therefore trigger an inflammatory response upon attaching to TLRs. TLR-2 present at the endometrial glands is important because it removes ‘less fit’ and excess spermatozoa, possibly to prevent polyspermy, as well as prepare the uterus for impending nidation [47]. Postpartum, uterine infections are known to disrupt endometrial epithelial integrity and reduce uterine gland functions by disrupting prostaglandin secretion, thereby compromising folliculogenesis and perturbing fertility [40,48,49]. Many reproductive infections are known to impair fertility. If present in the uterus, E. coli, T. pyogenes exotoxin, and Bovine Herpesvirus 4 (BoHV-4) are known to disrupt reproductive hormone secretion [43,50,51]. For instance, E. coli secretes LPS, which increases prostaglandin E2 (PGE2) production by the endometrial glands instead of PGF2α [43]. PGF2α is luteolytic hormone, while PGE2 is a luteotropic hormone that causes persistence of the corpus luteum in absence of a conception, resulting in pseudopregnancy or luteal cysts and anestrus. In addition, progesterone produced by the persistent corpus luteum dampens immune responses, facilitates pathogen proliferation, and impairs fertility [52,53]. The highly pathogenic bacteria T. pyogenes produces an aggressive toxin called pyolysin which damages endometrial epithelial cells, leading to dysregulated hormonal secretion and disrupted fertility [28]. Furthermore, low counts of Corynebacterium, Staphylococcus, and Prevotella two days before insemination are known to improve pregnancy rates in cattle [21].
While the beneficial effects of intravaginal Lactobacillus are not yet well known, in the gut, it produces phenolics which protect the oocyte against oxidative stress, thus improving oocyte competency and fertilization [54,55,56]. Infusing Lactobacillus as prebiotics during cattle breeding would likely improve fertility when better quality oocytes are ovulated and fertilized [54,57]. In addition, Lactobacillus suppress the production of eicosanoids, which are implicated in the inflammatory reaction and negatively affect the oocyte quality when produced at high concentrations. In humans, Lactobacillus delbrueckii is abundant in the vagina and is known to increase sperm capacitation through the production of bicarbonate ion (HCO3) from water and carbon dioxide [58,59]. To improve fertility in cattle, further studies on the benefits of using such probiotics around the time of breeding are warranted. This may result in reduction in number of services per conception and increased conception to first service and higher pregnancy rates.

5. Genital Microbiome and Pregnancy

Technologies such as fluorescence in situ hybridization and 16s rRNA gene sequencing allowed the identification of bacterial species in pregnant cattle [11,60]. The detected bacteria are often associated with uterine diseases and abortion; however, the pregnant cows were clinically healthy. Thus, the gravid uterus is not sterile, contrary to what was previously thought [61]. Interestingly, the abundance and diversity of the microbiomes differ widely from that found in diseased endometria. The microbial load and diversity within the genital tract during pregnancy are relatively low, possibly to reduce the risk of dysbiosis and abortion [1,62]. The common bacterial phyla found in the pregnant uterus of cattle, at placentome and inter-cotyledonary placenta, are Firmicutes and Bacteroidetes [11]. Another study, using fluorescence in situ hybridization, detected an abundance of F. necrophorum, T. pyogenes, and Porphyromonas levii in the endometrium and placentomes of cows [60]. It seems that the presence of Lactobacillus is beneficial for the success of pregnancy. In humans, the presence of Lactobacillus was associated with good placental growth and angiogenesis [63]. In addition, Lactobacillus decreases the production of proinflammatory cytokines, which improves the immune tolerance of the conceptus and facilitates the nidation process [64]. Same statements were reported for in vitro fertilization where the dominance of the non-Lactobacillus microbiome on the endometrium was associated with decreased implantation rate [63]. In pregnant women, the absence of or decrease in Lactobacillus population in the vagina reduces the competition effect against pathogens, leading to an overgrowth of more invasive microbes which will trigger an inflammatory reaction associated with the high release of cytokines, mainly Interleukin (IL)-8, and premature delivery or abortion [65]. Within this context, bovine endometrial epithelial cells were cultured with four different Lactobacillus strains elaborated with proinflammatory cytokines IL 1A, IL6 and IL8 [66]. The study confirmed that some Lactobacillus strains, such as Lactobacillus ruminis and Lactobacillus amylovorus, increased the cytokine gene expression concomitantly with the increased microbial load. It is possible that low cytokine production stimulated by Lactobacillus has immunomodulatory effects. Without Lactobacillus, pathogens proliferate, stimulating excessive proinflammatory responses and thereby perturbing fertility. On the other hand, a high abundance of S. aureus and T. pyogenes in the bovine genital tract were associated with an increased risk of abortion [67,68]. Therefore, dysbiosis of the vaginal and endometrial microbiomes hinders fertility.

6. Genital Microbiome and Postpartum Complications

The postpartum period is a critical window of the production cycle of cattle. Perturbed uterine involution can result in metritis, endometritis, or cystic ovarian disease. To optimize fertility, uterine involution should be uninterrupted and completed around 45 days postpartum. However, bacteria that contaminate the uterus during parturition could slow these two events [69,70,71]. These bacteria are originated from the external environment or the animal’s adjacent organs [11,15,60]. Retained fetal membranes (RFM), metritis, and endometritis are the most frequent postpartum complications in cattle. In cows with normal pregnancy and parturition, all females have a similar microbiome at calving that then becomes divergent by seven days postpartum (DPP), when Fusobacteria and Bacteroidetes predominate in cows that experienced dystocia or RFM [71]. It is thought that the genital tract microbiome during the postpartum period has a profound effect on subsequent reproductive performances [22,72]. RFM is the persistence of placental tissue attached to the endometrium, representing a favorable environment for the growth of pathogenic bacteria, leading to subfertility [73]. Using culture- and molecular-based methods for microbiology identification, E. coli (68%) and S. aureus (18%) were the most associated to RFM [74]. Furthermore, RFM is an important risk factor for uterine diseases, mainly metritis and endometritis [73,75,76].
Metritis is a deep inflammation of the endometrium and myometrium, which is associated with purulent and or fetid vaginal discharge, detected before day 21 postpartum [77]. Cows with metritis harbor a less diverse microbiome dominated by Bacteroides, Porphyromonas, and Fusobacterium [5,78]. It is thought that the higher abundance of Proteobacteria in the vagina seven days before calving predicts the occurrence of metritis postpartum, because it synergizes Fusobacteria [71]. E. coli is highly detected in uterine samples in the case of metritis, and the severity of symptoms is more important when the disease is induced by E. coli strains harboring the virulence factor kpsMTII [79]. In addition, a symbiotic relationship between E. coli, T. pyogenes, and F. necrophorum facilitates colonization of the endometrium and evasion of the immune system, leading to metritis [80]. T. pyogenes produce a powerful cytolytic substance, pyolysin, which alters the endometrial epithelial cell membranes, leading to a tissue damage and disruption of the mucosal integrity [28]. While the endometrial cells do not respond to damaged-associated molecular pattern (DAMP), the combination of pathogens and DAMP triggers an inflammatory reaction through the intracellular secretion of IL-1 by the endometrial cells [28].
Endometritis is a superficial inflammation of the endometrium, which is associated with less severe clinical symptoms compared to metritis and detected after day 21 postpartum, as it is considered as abnormally sustained postpartum inflammation [77,81]. Endometritis is highly debated regarding the origin and pathogenesis of the disease. While it is not confirmed that endometritis is triggered by bacteria, rather than uncontrolled uterine inflammation, several microbes were isolated in animals with clinical and subclinical endometritis [28,29,30,72]. Metagenomic analyses of 16S rRNA gene sequences of the uterine microbiome in endometrial cytobrush collected at different times during the first week postpartum revealed that metritis and clinical endometritis are associated with a lower microbial diversity postpartum, dominated by Bacteroidetes, Fusobacterium, and Trueperella, and lower abundance of Escherichia, Shigella, Lactobacillus, Prevotella, Schlegelella, and Streptococcus [28,29,30,71]. In addition, abundant Anaerococcus, Corynebacterium, and Staphylococcus increase the risk of subclinical endometritis in postpartum cows [28,29,30]. An interesting synergy between T. pyogenes, F. necrophorum, and Prevotella species would affect the severity of the clinical symptoms of endometritis [82].
Interestingly, the vaginal and uterine microbiome during the postpartum period affect the reproductive performances in subsequent breeding [22]. For instance, gene sequencing targeting the V1 to V3 hypervariable regions of the 16S rRNA bacterial gene in uterine and vaginal flushes confirmed that low abundance of Corynebacterium, Staphylococcus, and Prevotella two days before insemination improve the pregnancy rate [21]. Therefore, it is important to control the genital microbiome before insemination using probiotics to improve fertility.

7. Genital Microbiome and Animal Behavior

The microbiome of different organs confirmed to be effective in modulating the animal behavior toward its conspecifics. The most extensive studies regarding pheromone production are conducted in wild animals [83,84,85]. However, it is possible that microbiomes of livestock, including cows, may show similar diversity patterns. It is thought that microbiomes and animal behavior in wild mammals mirror ruminants [26]. For instance, the microbiome and volatile secretions of the anal scent gland were compared between spotted and stripped hyenas [85]. Interestingly, secretions of the scent gland vary according to the detected microbiome. In addition, both volatile and microbiome profiles of the anal scent gland were different between spotted and stripped hyenas and vary according to the reproductive status of the animal, all of which means that the microbiome and by-products of volatile molecules are highly specific to the species and social groups. The same findings were reported in wild meerkats [86].
Likewise, trimethylamine is a powerful volatile substance produced by mice and helps in matting. Interestingly, trimethylamine is highly specific to the species, as it attracts mice but repels rats. Specific commensal bacteria of the intestine metabolize the dietary choline to produce trimethylamine [3]. Likewise, the genital microbiome of different animals, including bovines, was confirmed to be implicated in pheromone production and semiochemical signaling between females and males [87]. This is a solid confirmation that any changes in the female’s genital microbiome affect her pheromone profile and other males’ behavior toward the female. Thus, the genital microbiome of an animal does not only modify the organism’s function and volatile profile but also affects the behavior of the other animals, including predator–prey interaction and feeding behavior [88,89]. Likewise, the females of Anopheles gambiae, the African malaria mosquito, target their hosts based on the chemical cues released that they produce and the load and diversity of the skin microbiome. Humans with low skin microbial diversity are the target of choice for mosquitos. In addition, the presence of some bacteria (i.e., Pseudomonas spp.) repels the mosquitos [88].
Bovine cervicovaginal mucus (CVM) contains abundant commensal microbes and volatile compounds. During estrus, CVM is highly rich in pheromones that are specific to estrus, among which oleic acid, trimethylamine, acetic acid, and propionic acid are the most important [27]. It seems that pheromones are more concentrated in the CVM than any other body secretions. In an interesting experiment, urine, saliva, feces, and CVM of females in estrus were rubbed on the vulva of anestrus females; these females were exposed to males and Flehmen behavior was monitored [87]. Secretions of other females at pre-estrus and post-estrus were also compared, and water was used as a negative control. Interestingly, the CVM resulted in the highest and longest Flehmen behavior compared to urine, saliva, and feces. No Flehmen reaction was reported in the secretions of females that were not in estrus. Furthermore, the CVM of buffalo during estrus is highly rich in Firmicutes, which are fermentative anaerobic bacteria [23]. Firmicutes, the phylum which includes the Lactobacillales order, is abundant in the genital tract of healthy cows compared to those with uterine disease [5,90]. These bacteria are well known to be implicated in the pheromone production in anal scent glands of hyena [85]. Any modification of the genital microbiome by external factors would affect the volatile profile of CVM. Likewise, intravaginal administration of antibiotics in bitches at estrus reduces the attractiveness of males compared to the untreated females [91]. These findings confirm that pheromones are highly produced in the genital tract and are implicated in the sexual communication between males and females. Thus, it is important to reduce antibiotic use intravaginally, since it affects the microbial diversity of the genital tract and the produced pheromones.

8. Genital Tract Microbiome and Probiotics

Metritis is caused by several bacteria, mainly E. coli, F. necrophorum, and T. pyogenes [5,77,80]. Due to the high economic loss associated with medication, milk withdrawal, and reduced fertility, antibiotics and antimicrobials are often used to control the disease. Third-generation cephalosporins are the molecule of choice for the treatment of metritis. However, many countries such as the USA have banned their use in food animals, to be only reserved for human medicine [92]. In addition, abusive use of antibiotics increased the risk of the development of resistant strains and the accumulation of antibiotic residues in milk and meat [93,94]. Therefore, developing new alternative protocols for a better management of reproductive diseases is a must. The term “probiotics” represents live microorganisms that confer health benefits on the host when they are administered in adequate amounts [95]. The use of probiotics is promising in treating human urogenital diseases. Lactobacillus maybe be part of a bacterial resident population in the uterus constituting a protective microbiome. Thus, uterine microbiota and specific bacterial species may be linked to critical health status such as endometritis, improved conception rates, or abortion. Cattle fertility will benefit from expanded studies of genital tract microbiota interactions. Lactobacillus are widely used as probiotics for treating different genital or general diseases because they seem to be tolerated by the host. In addition, Lactobacillus spp. can modulate the immunity of the host by producing bioactive molecules such as lactic acid and hydrogen peroxide [9,10]. Finally, it seems that these bacteria are highly tolerated by the host organism and are rarely implicated in genital issues. For example, Lactobacillus was used to treat vulvovaginal candidiasis, a fungal infestation of the vagina induced by Candida albicans [96]. The patients received capsules of Lactobacillus plantarum intravaginally for five weeks, which resulted in better resolution of symptoms than in the patients that received clotrimazole alone as intravaginal cream. Likewise, Lactobacillus crispatus were successfully used to manage recurrent urinary tract infections, mainly induced by E. coli [97]. The patients were treated with intravaginal capsules containing Lactobacillus crispatus for 15 days, which resulted in a lower recurrency rate of the disease compared to the control group.
Furthermore, oral supplementation of mice with Lactobaccillus rhamnosus resulted in better swimming test, lower anxiety, and higher expression of γ-aminobutyric acid (GABA) receptors in the brain [98]. GABA is a powerful inhibitory neurotransmitter in the central nervous system that reduces neuronal excitability throughout the nervous system [99]. The partial removal of the vagus nerve, which communicates between the gastro-intestinal tract and the brain, resulted in an obliteration of the effects induced by Lactobacillus supplementation. This is a clear statement that all the information of the digestive system including the gut microbiome is transmitted to the brain and affects the animal’s behavior.
Lactobacillus would likely to play a significant role in controlling the postpartum uterine diseases when infused directly intravaginally as probiotics. Incidences of endometrial inflammation postpartum were reduced when Lactobacillus sakei and Pediococcus acidilactici were introduced intravaginally in animals before and after calving [100,101]. Pediococcus are another valuable probiotic, as they have specific genes encoding for Pediocin, a potent bactericidal peptide [102,103]. Likewise, Lactobacillus buchneri were introduced intravaginally in dairy cows between 24 and 30 DPP, and the uterine health status and reproductive performances were measured and compared to a control group [94]. Probiotic treatment resulted in shorter median days to first service, lower number of services per conception, higher first service conception rate, and shorter median days to conception compared to the placebo group treated with isotonic saline solution. In addition, gene expression of pro-inflammatory cytokines and chemokines was lower in the probiotic group. These results confirm that the presence of Lactobacillus in the genital tract protects the genital health status and improves fertility of cows.

9. Genital Tract Microbiome and Prebiotics

Alternatively, favorable conditions that encourage the proliferation of a healthy microbiome as prebiotics can be ensured [61,104]. Providing a high-energy diet around calving affects the diversity of the uterine microbiome and the occurrence of uterine inflammation. For instance, pregnant multiparous Holstein cows were fed with a diet that covered 80% of the energy requirement, while the other group received a diet that covered 100% of the energy requirement, between 20 days pre-calving and 30 days postpartum [104]. As major results, the energy restriction resulted in an abundance of Bacteroidetes and Fusobacteria phyla and higher expression of inflammatory cytokines in the endometrium. According to a previous study, to switch from a healthy uterus postpartum to a metritic uterus, there must be an overgrowth of Bacteroidetes and Fusobacteria and reduced abundance of Proteobacteria and Tenericutes [5].

10. Conclusions

Various organs have distinct microbial inhabitants. Apart from the gut, the group that has attracted the most attention in livestock research is the one in the genital tract. Altogether, these examples indicate the involvement of genital tract bacteria in improving fertility. While the consequences of reducing bacterial diversity in the genital tract microbiome remain unexplored, intravaginal inoculation of cattle at risk of infection represents possible probiotic management of postpartum uterine diseases. It maybe that probiotics that alter the endometrial microbiome could provide viable alternatives to existing therapies for uterine disease in cattle.

Author Contributions

All the authors state that they contributed equally to writing and editing of the paper in its final form and approved it for submission. The authors state that the manuscript has not been previously published and that the manuscript is not under consideration elsewhere. All authors have read and agreed to the published version of the manuscript.

Funding

This review did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. Publication fees were provided by the Center for One Health (C1), Ross University School of Veterinary Medicine.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Laguardia-Nascimento, M.; Branco, K.M.; Gasparini, M.R.; Giannattasio-Ferraz, S.; Leite, L.R.; Araujo, F.M.; Salim, A.C.; Nicoli, J.R.; de Oliveira, G.C.; Barbosa-Stancioli, E.F. Vaginal Microbiome Characterization of Nellore Cattle Using Metagenomic Analysis. PLoS ONE 2015, 10, e0143294. [Google Scholar] [CrossRef] [PubMed]
  2. Owens, C.E.; Daniels, K.M.; Ealy, A.D.; Knowlton, K.F.; Cockrum, R.R. Graduate Student Literature Review: Potential mechanisms of interaction between bacteria and the reproductive tract of dairy cattle. J. Dairy Sci. 2020, 103, 10951–10960. [Google Scholar] [CrossRef] [PubMed]
  3. Al-Waiz, M.; Mikov, M.; Mitchell, S.C.; Smith, R.L. The exogenous origin of trimethylamine in the mouse. Metabolism 1992, 41, 135–136. [Google Scholar] [CrossRef]
  4. Xu, Q.; Qiao, Q.; Gao, Y.; Hou, J.; Hu, M.; Du, Y.; Zhao, K.; Li, X. Gut Microbiota and Their Role in Health and Metabolic Disease of Dairy Cow. Front Nutr 2021, 8, 701511. [Google Scholar] [CrossRef] [PubMed]
  5. Galvao, K.N.; Bicalho, R.C.; Jeon, S.J. Symposium review: The uterine microbiome associated with the development of uterine disease in dairy cows. J. Dairy Sci. 2019, 102, 11786–11797. [Google Scholar] [CrossRef]
  6. Sheldon, I.M.; Cronin, J.G.; Bromfield, J.J. Tolerance and Innate Immunity Shape the Development of Postpartum Uterine Disease and the Impact of Endometritis in Dairy Cattle. Annu. Rev. Anim. Biosci. 2019, 7, 361–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Kovachev, S. Defence factors of vaginal lactobacilli. Crit. Rev. Microbiol. 2018, 44, 31–39. [Google Scholar] [CrossRef]
  8. Punzon-Jimenez, P.; Labarta, E. The impact of the female genital tract microbiome in women health and reproduction: A review. J. Assist. Reprod. Genet. 2021, 38, 2519–2541. [Google Scholar] [CrossRef]
  9. Tachedjian, G.; Aldunate, M.; Bradshaw, C.S.; Cone, R.A. The role of lactic acid production by probiotic Lactobacillus species in vaginal health. Res. Microbiol. 2017, 168, 782–792. [Google Scholar] [CrossRef]
  10. Martin, R.; Suarez, J.E. Biosynthesis and degradation of H2O2 by vaginal lactobacilli. Appl. Environ. Microbiol. 2010, 76, 400–405. [Google Scholar] [CrossRef]
  11. Moore, S.G.; Ericsson, A.C.; Poock, S.E.; Melendez, P.; Lucy, M.C. Hot topic: 16S rRNA gene sequencing reveals the microbiome of the virgin and pregnant bovine uterus. J. Dairy Sci. 2017, 100, 4953–4960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Adnane, M.; Chapwanya, A. A Review of the Diversity of the Genital Tract Microbiome and Implications for Fertility of Cattle. Animals 2022, 12, 460. [Google Scholar] [CrossRef] [PubMed]
  13. Schuenemann, G.M.; Bas, S.; Gordon, E.; Workman, J.D. Dairy calving management: Description and assessment of a training program for dairy personnel. J. Dairy Sci. 2013, 96, 2671–2680. [Google Scholar] [CrossRef] [PubMed]
  14. Swartz, J.D.; Lachman, M.; Westveer, K.; O’Neill, T.; Geary, T.; Kott, R.W.; Berardinelli, J.G.; Hatfield, P.G.; Thomson, J.M.; Roberts, A.; et al. Characterization of the Vaginal Microbiota of Ewes and Cows Reveals a Unique Microbiota with Low Levels of Lactobacilli and Near-Neutral pH. Front. Vet. Sci. 2014, 1, 19. [Google Scholar] [CrossRef] [Green Version]
  15. Ong, C.T.; Turni, C.; Blackall, P.J.; Boe-Hansen, G.; Hayes, B.J.; Tabor, A.E. Interrogating the bovine reproductive tract metagenomes using culture-independent approaches: A systematic review. Anim. Microbiome 2021, 3, 41. [Google Scholar] [CrossRef]
  16. Rodríguez, C.; Cofré, J.V.; Sánchez, M.; Fernández, P.; Boggiano, G.; Castro, E. Lactobacilli isolated from vaginal vault of dairy and meat cows during progesteronic stage of estrous cycle. Anaerobe 2011, 17, 15–18. [Google Scholar] [CrossRef]
  17. O’Hanlon, D.E.; Moench, T.R.; Cone, R.A. Vaginal pH and microbicidal lactic acid when lactobacilli dominate the microbiota. PLoS ONE 2013, 8, e80074. [Google Scholar] [CrossRef]
  18. Nardini, P.; Nahui Palomino, R.A.; Parolin, C.; Laghi, L.; Foschi, C.; Cevenini, R.; Vitali, B.; Marangoni, A. Lactobacillus crispatus inhibits the infectivity of Chlamydia trachomatis elementary bodies, in vitro study. Sci. Rep. 2016, 6, 29024. [Google Scholar] [CrossRef] [Green Version]
  19. Graver, M.A.; Wade, J.J. The role of acidification in the inhibition of Neisseria gonorrhoeae by vaginal lactobacilli during anaerobic growth. Ann. Clin. Microbiol. Antimicrob. 2011, 10, 8. [Google Scholar] [CrossRef] [Green Version]
  20. Valore, E.V.; Park, C.H.; Igreti, S.L.; Ganz, T. Antimicrobial components of vaginal fluid. Am. J. Obstet. Gynecol. 2002, 187, 561–568. [Google Scholar] [CrossRef]
  21. Ault, T.B.; Clemmons, B.A.; Reese, S.T.; Dantas, F.G.; Franco, G.A.; Smith, T.P.L.; Edwards, J.L.; Myer, P.R.; Pohler, K.G. Bacterial taxonomic composition of the postpartum cow uterus and vagina prior to artificial insemination. J. Anim. Sci. 2019, 97, 4305–4313. [Google Scholar] [CrossRef]
  22. Ault, T.B.; Clemmons, B.A.; Reese, S.T.; Dantas, F.G.; Franco, G.A.; Smith, T.P.L.; Edwards, J.L.; Myer, P.R.; Pohler, K.G. Uterine and vaginal bacterial community diversity prior to artificial insemination between pregnant and nonpregnant postpartum cows1. J. Anim. Sci. 2019, 97, 4298–4304. [Google Scholar] [CrossRef] [PubMed]
  23. Mahalingam, S.; Dharumadurai, D.; Archunan, G. Vaginal microbiome analysis of buffalo (Bubalus bubalis) during estrous cycle using high-throughput amplicon sequence of 16S rRNA gene. Symbiosis 2019, 78, 97–106. [Google Scholar] [CrossRef]
  24. Vitale, S.G.; Ferrari, F.; Ciebiera, M.; Zgliczynska, M.; Rapisarda, A.M.C.; Vecchio, G.M.; Pino, A.; Angelico, G.; Knafel, A.; Riemma, G.; et al. The Role of Genital Tract Microbiome in Fertility: A Systematic Review. Int. J. Mol. Sci. 2021, 23, 180. [Google Scholar] [CrossRef]
  25. Agarwal, K.; Lewis, A.L. Vaginal sialoglycan foraging by Gardnerella vaginalis: Mucus barriers as a meal for unwelcome guests? Glycobiology 2021, 31, 667–680. [Google Scholar] [CrossRef]
  26. Srinivasan, M.; Adnane, M.; Archunan, G. Significance of cervico-vaginal microbes in bovine reproduction and pheromone production—A hypothetical review. Res. Vet. Sci. 2021, 135, 66–71. [Google Scholar] [CrossRef] [PubMed]
  27. Archunan, G.; Rajanarayanan, S.; Karthikeyan, K. Cattle Pheromones. In Neurobiology of Chemical Communication; Mucignat-Caretta, C., Ed.; CRC Press: Boca Raton, FL, USA; Taylor & Francis: Abingdon, UK, 2014. [Google Scholar]
  28. Carneiro, L.C.; Cronin, J.G.; Sheldon, I.M. Mechanisms linking bacterial infections of the bovine endometrium to disease and infertility. Reprod. Biol. 2016, 16, 1–7. [Google Scholar] [CrossRef] [Green Version]
  29. Pascottini, O.B.; Van Schyndel, S.J.; Spricigo, J.F.W.; Rousseau, J.; Weese, J.S.; LeBlanc, S.J. Dynamics of uterine microbiota in postpartum dairy cows with clinical or subclinical endometritis. Sci. Rep. 2020, 10, 12353. [Google Scholar] [CrossRef]
  30. Miranda-CasoLuengo, R.; Lu, J.; Williams, E.J.; Miranda-CasoLuengo, A.A.; Carrington, S.D.; Evans, A.C.O.; Meijer, W.G. Delayed differentiation of vaginal and uterine microbiomes in dairy cows developing postpartum endometritis. PLoS ONE 2019, 14, e0200974. [Google Scholar] [CrossRef] [Green Version]
  31. Liu, M.; Wu, Q.; Wang, M.; Fu, Y.; Wang, J. Lactobacillus rhamnosus GR-1 Limits Escherichia coli-Induced Inflammatory Responses via Attenuating MyD88-Dependent and MyD88-Independent Pathway Activation in Bovine Endometrial Epithelial Cells. Inflammation 2016, 39, 1483–1494. [Google Scholar] [CrossRef]
  32. Genis, S.; Sanchez-Chardi, A.; Bach, A.; Fabregas, F.; Aris, A. A combination of lactic acid bacteria regulates Escherichia coli infection and inflammation of the bovine endometrium. J. Dairy Sci. 2017, 100, 479–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Otero, M.C.; Nader-Macias, M.E. Inhibition of Staphylococcus aureus by H2O2-producing Lactobacillus gasseri isolated from the vaginal tract of cattle. Anim. Reprod. Sci. 2006, 96, 35–46. [Google Scholar] [CrossRef] [PubMed]
  34. Otero, M.C.; Morelli, L.; Nader-Macias, M.E.F. Probiotic properties of vaginal lactic acid bacteria to prevent metritis in cattle. Lett. Appl. Microbiol. 2006, 43, 91–97. [Google Scholar] [CrossRef] [PubMed]
  35. Alakomi, H.L.; Skytta, E.; Saarela, M.; Mattila-Sandholm, T.; Latva-Kala, K.; Helander, I.M. Lactic acid permeabilizes gram-negative bacteria by disrupting the outer membrane. Appl. Environ. Microbiol. 2000, 66, 2001–2005. [Google Scholar] [CrossRef] [Green Version]
  36. Wang, J.; Liu, C.; Nesengani, L.T.; Gong, Y.; Yang, Y.; Yang, L.; Lu, W. Comparison of vaginal microbial community structure of beef cattle between luteal phase and follicular phase. Indian J. Anim. Res. 2019, 53, 1298–1303. [Google Scholar] [CrossRef]
  37. Moore, S.G.; Ericsson, A.C.; Behura, S.K.; Lamberson, W.R.; Evans, T.J.; McCabe, M.S.; Poock, S.E.; Lucy, M.C. Concurrent and long-term associations between the endometrial microbiota and endometrial transcriptome in postpartum dairy cows. BMC Genom. 2019, 20, 405. [Google Scholar] [CrossRef]
  38. Perry, G.A.; Perry, B.L. Effect of preovulatory concentrations of estradiol and initiation of standing estrus on uterine pH in beef cows. Domest. Anim. Endocrinol. 2008, 34, 333–338. [Google Scholar] [CrossRef]
  39. Messman, R.D.; Contreras-Correa, Z.E.; Paz, H.A.; Perry, G.; Lemley, C.O. Vaginal bacterial community composition and concentrations of estradiol at the time of artificial insemination in Brangus heifers. J. Anim. Sci. 2020, 98, skaa178. [Google Scholar] [CrossRef]
  40. Sheldon, I.M.; Price, S.B.; Cronin, J.; Gilbert, R.O.; Gadsby, J.E. Mechanisms of infertility associated with clinical and subclinical endometritis in high producing dairy cattle. Reprod. Domest. Anim.—Zuchthygiene 2009, 44 (Suppl 3), 1–9. [Google Scholar] [CrossRef]
  41. Sheldon, I.M.; Noakes, D.E.; Rycroft, A.N.; Pfeiffer, D.U.; Dobson, H. Influence of uterine bacterial contamination after parturition on ovarian dominant follicle selection and follicle growth and function in cattle. Reproduction 2002, 123, 837–845. [Google Scholar] [CrossRef]
  42. Cronin, J.G.; Turner, M.L.; Goetze, L.; Bryant, C.E.; Sheldon, I.M. Toll-like receptor 4 and MYD88-dependent signaling mechanisms of the innate immune system are essential for the response to lipopolysaccharide by epithelial and stromal cells of the bovine endometrium. Biol. Reprod. 2012, 86, 51. [Google Scholar] [CrossRef] [PubMed]
  43. Herath, S.; Lilly, S.T.; Fischer, D.P.; Williams, E.J.; Dobson, H.; Bryant, C.E.; Sheldon, I.M. Bacterial lipopolysaccharide induces an endocrine switch from prostaglandin F2alpha to prostaglandin E2 in bovine endometrium. Endocrinology 2009, 150, 1912–1920. [Google Scholar] [CrossRef] [PubMed]
  44. Herath, S.; Williams, E.J.; Lilly, S.T.; Gilbert, R.O.; Dobson, H.; Bryant, C.E.; Sheldon, I.M. Ovarian follicular cells have innate immune capabilities that modulate their endocrine function. Reproduction 2007, 134, 683–693. [Google Scholar] [CrossRef] [PubMed]
  45. Bromfield, J.J.; Santos, J.E.; Block, J.; Williams, R.S.; Sheldon, I.M. Physiology and Endocrinology Symposium: Uterine infection: Linking infection and innate immunity with infertility in the high-producing dairy cow. J. Anim. Sci. 2015, 93, 2021–2033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Williams, E.J.; Fischer, D.P.; Noakes, D.E.; England, G.C.; Rycroft, A.; Dobson, H.; Sheldon, I.M. The relationship between uterine pathogen growth density and ovarian function in the postpartum dairy cow. Theriogenology 2007, 68, 549–559. [Google Scholar] [CrossRef] [Green Version]
  47. Akthar, I.; Suarez, S.S.; Morillo, V.A.; Sasaki, M.; Ezz, M.A.; Takahashi, K.I.; Shimada, M.; Marey, M.A.; Miyamoto, A. Sperm enter glands of preovulatory bovine endometrial explants and initiate inflammation. Reproduction 2020, 159, 181–192. [Google Scholar] [CrossRef]
  48. Sheldon, I.M.; Cronin, J.; Goetze, L.; Donofrio, G.; Schuberth, H.J. Defining postpartum uterine disease and the mechanisms of infection and immunity in the female reproductive tract in cattle. Biol. Reprod. 2009, 81, 1025–1032. [Google Scholar] [CrossRef] [Green Version]
  49. Sicsic, R.; Goshen, T.; Dutta, R.; Kedem-Vaanunu, N.; Kaplan-Shabtai, V.; Pasternak, Z.; Gottlieb, Y.; Shpigel, N.Y.; Raz, T. Microbial communities and inflammatory response in the endometrium differ between normal and metritic dairy cows at 5–10 days post-partum. Vet. Res. 2018, 49, 77. [Google Scholar] [CrossRef] [Green Version]
  50. Donofrio, G.; Herath, S.; Sartori, C.; Cavirani, S.; Flammini, C.F.; Sheldon, I.M. Bovine herpesvirus 4 is tropic for bovine endometrial cells and modulates endocrine function. Reproduction 2007, 134, 183–197. [Google Scholar] [CrossRef]
  51. Davies, D.; Meade, K.G.; Herath, S.; Eckersall, P.D.; Gonzalez, D.; White, J.O.; Conlan, R.S.; O’Farrelly, C.; Sheldon, I.M. Toll-like receptor and antimicrobial peptide expression in the bovine endometrium. Reprod. Biol. Endocrinol. 2008, 6, 53. [Google Scholar] [CrossRef]
  52. Schust, D.J.; Anderson, D.J.; Hill, J.A. Progesterone-induced immunosuppression is not mediated through the progesterone receptor. Hum. Reprod. 1996, 11, 980–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Watson, E.D.; Stokes, C.R.; David, J.S.; Bourne, F.J. Effect of ovarian hormones on promotion of bactericidal activity by uterine secretions of ovariectomized mares. J. Reprod. Fertil. 1987, 79, 531–537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Guerreiro, T.M.; Gonçalves, R.F.; Melo, C.F.O.R.; de Oliveira, D.N.; Lima, E.d.O.; Visintin, J.A.; de Achilles, M.A.; Catharino, R.R. A Metabolomic Overview of Follicular Fluid in Cows. Front. Vet. Sci. 2018, 5, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Martins, N.; Barros, L.; Ferreira, I.C.F.R. In vivo antioxidant activity of phenolic compounds: Facts and gaps. Trends Food Sci. Technol. 2016, 48, 1–12. [Google Scholar] [CrossRef] [Green Version]
  56. Rodriguez, H.; Curiel, J.A.; Landete, J.M.; de las Rivas, B.; Lopez de Felipe, F.; Gomez-Cordoves, C.; Mancheno, J.M.; Munoz, R. Food phenolics and lactic acid bacteria. Int. J. Food Microbiol. 2009, 132, 79–90. [Google Scholar] [CrossRef] [Green Version]
  57. Jabbour, H.N.; Sales, K.J.; Catalano, R.D.; Norman, J.E. Inflammatory pathways in female reproductive health and disease. Reproduction 2009, 138, 903–919. [Google Scholar] [CrossRef] [PubMed]
  58. Li, C.-X.; Jiang, X.-C.; Qiu, Y.-J.; Xu, J.-H. Identification of a new thermostable and alkali-tolerant α-carbonic anhydrase from Lactobacillus delbrueckii as a biocatalyst for CO2 biomineralization. Bioresour. Bioprocess. 2015, 2, 44. [Google Scholar] [CrossRef] [Green Version]
  59. Visconti, P.E.; Krapf, D.; de la Vega-Beltran, J.L.; Acevedo, J.J.; Darszon, A. Ion channels, phosphorylation and mammalian sperm capacitation. Asian J. Androl. 2011, 13, 395–405. [Google Scholar] [CrossRef] [Green Version]
  60. Karstrup, C.C.; Klitgaard, K.; Jensen, T.K.; Agerholm, J.S.; Pedersen, H.G. Presence of bacteria in the endometrium and placentomes of pregnant cows. Theriogenology 2017, 99, 41–47. [Google Scholar] [CrossRef]
  61. Fulop, V.; Demeter, J.; Cseh, A. Significance and effects of prenatal and postnatal microbiome in the period of early individual development and options for interventional treatment. Orv. Hetil. 2021, 162, 731–740. [Google Scholar] [CrossRef]
  62. Deng, F.; McClure, M.; Rorie, R.; Wang, X.; Chai, J.; Wei, X.; Lai, S.; Zhao, J. The vaginal and fecal microbiomes are related to pregnancy status in beef heifers. J. Anim. Sci. Biotechnol. 2019, 10, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Moreno, I.; Codoner, F.M.; Vilella, F.; Valbuena, D.; Martinez-Blanch, J.F.; Jimenez-Almazan, J.; Alonso, R.; Alama, P.; Remohi, J.; Pellicer, A.; et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am. J. Obstet. Gynecol. 2016, 215, 684–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Vacca, P.; Cantoni, C.; Vitale, M.; Prato, C.; Canegallo, F.; Fenoglio, D.; Ragni, N.; Moretta, L.; Mingari, M.C. Crosstalk between decidual NK and CD14+ myelomonocytic cells results in induction of Tregs and immunosuppression. Proc. Natl. Acad. Sci. USA 2010, 107, 11918–11923. [Google Scholar] [CrossRef] [Green Version]
  65. Sakai, M.; Ishiyama, A.; Tabata, M.; Sasaki, Y.; Yoneda, S.; Shiozaki, A.; Saito, S. Relationship between cervical mucus interleukin-8 concentrations and vaginal bacteria in pregnancy. Am. J. Reprod. Immunol. 2004, 52, 106–112. [Google Scholar] [CrossRef]
  66. Gartner, M.A.; Bondzio, A.; Braun, N.; Jung, M.; Einspanier, R.; Gabler, C. Detection and characterisation of Lactobacillus spp. in the bovine uterus and their influence on bovine endometrial epithelial cells in vitro. PLoS ONE 2015, 10, e0119793. [Google Scholar] [CrossRef] [Green Version]
  67. Galvão, K. Postpartum uterine diseases in dairy cows. Proc. Anim. Reprod. 2012, 9, 290–296. [Google Scholar]
  68. Anderson, M.L. Infectious causes of bovine abortion during mid- to late-gestation. Theriogenology 2007, 68, 474–486. [Google Scholar] [CrossRef]
  69. Peng, Y.; Wang, Y.; Hang, S.; Zhu, W. Microbial diversity in uterus of healthy and metritic postpartum Holstein dairy cows. Folia Microbiol. 2013, 58, 593–600. [Google Scholar] [CrossRef]
  70. Elkjaer, K.; Ancker, M.L.; Gustafsson, H.; Friggens, N.C.; Waldmann, A.; Molbak, L.; Callesen, H. Uterine bacterial flora in postpartum Danish Holstein dairy cows determined using DNA-based fingerprinting: Correlation to uterine condition and calving management. Anim. Reprod. Sci. 2013, 138, 39–48. [Google Scholar] [CrossRef] [Green Version]
  71. Bicalho, M.L.S.; Santin, T.; Rodrigues, M.X.; Marques, C.E.; Lima, S.F.; Bicalho, R.C. Dynamics of the microbiota found in the vaginas of dairy cows during the transition period: Associations with uterine diseases and reproductive outcome. J. Dairy Sci. 2017, 100, 3043–3058. [Google Scholar] [CrossRef]
  72. Wang, M.L.; Liu, M.C.; Xu, J.; An, L.G.; Wang, J.F.; Zhu, Y.H. Uterine Microbiota of Dairy Cows With Clinical and Subclinical Endometritis. Front. Microbiol. 2018, 9, 2691. [Google Scholar] [CrossRef] [PubMed]
  73. Potter, T.J.; Guitian, J.; Fishwick, J.; Gordon, P.J.; Sheldon, I.M. Risk factors for clinical endometritis in postpartum dairy cattle. Theriogenology 2010, 74, 127–134. [Google Scholar] [CrossRef] [PubMed]
  74. Al-Zubaidi, S.F.A.; Hasson, S.O.; Ajeel, H.H. Isolation and identification of the bacteria associated with the retained fetal membranes in cows by PCR technique in Babylon-Iraq. Euphrates J. Agric. Sci. 2017, 9, 18–24. [Google Scholar]
  75. Kim, I.H.; Kang, H.G. Risk factors for postpartum endometritis and the effect of endometritis on reproductive performance in dairy cows in Korea. J. Reprod. Dev. 2003, 49, 485–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Adnane, M.; Kaidi, R.; Hanzen, C.; England, G.C.W. Risk factors of clinical and subclinical endometritis in cattle: A review. Turk. J. Vet. Anim. Sci. 2017, 41, 1–11. [Google Scholar] [CrossRef]
  77. Sheldon, I.M.; Lewis, G.S.; LeBlanc, S.; Gilbert, R.O. Defining postpartum uterine disease in cattle. Theriogenology 2006, 65, 1516–1530. [Google Scholar] [CrossRef]
  78. Sheldon, I.M.; Williams, E.J.; Miller, A.N.; Nash, D.M.; Herath, S. Uterine diseases in cattle after parturition. Vet. J. 2008, 176, 115–121. [Google Scholar] [CrossRef]
  79. Yamamura, F.; Sugiura, T.; Munby, M.; Shiokura, Y.; Murata, R.; Nakamura, T.; Fujiki, J.; Iwano, H. Relationship between Escherichia coli virulence factors, notably kpsMTII, and symptoms of clinical metritis and endometritis in dairy cows. J. Vet. Med. Sci. 2022, 84, 420–428. [Google Scholar] [CrossRef]
  80. Bicalho, M.L.; Machado, V.S.; Oikonomou, G.; Gilbert, R.O.; Bicalho, R.C. Association between virulence factors of Escherichia coli, Fusobacterium necrophorum, and Arcanobacterium pyogenes and uterine diseases of dairy cows. Vet. Microbiol. 2012, 157, 125–131. [Google Scholar] [CrossRef]
  81. LeBlanc, S.J.; Duffield, T.F.; Leslie, K.E.; Bateman, K.G.; Keefe, G.P.; Walton, J.S.; Johnson, W.H. Defining and diagnosing postpartum clinical endometritis and its impact on reproductive performance in dairy cows. J. Dairy Sci. 2002, 85, 2223–2236. [Google Scholar] [CrossRef] [Green Version]
  82. Ruder, C.A.; Sasser, R.G.; Williams, R.J.; Ely, J.K.; Bull, R.C.; Butler, J.E. Uterine infections in the postpartum cow. Theriogenology 1981, 15, 573–580. [Google Scholar] [CrossRef]
  83. Hofer, H.; East, M.L.; Sämmang, I.; Dehnhard, M. Analysis of Volatile Compounds in Scent-Marks of Spotted Hyenas (Crocuta Crocuta) and their Possible Function in Olfactory Communication. In Chemical Signals in Vertebrates 9; Springer: Berlin/Heidelberg, Germany, 2001; pp. 141–148. [Google Scholar]
  84. Archie, E.A.; Theis, K.R. Animal behaviour meets microbial ecology. Anim. Behav. 2011, 82, 425–436. [Google Scholar] [CrossRef]
  85. Theis, K.R.; Venkataraman, A.; Dycus, J.A.; Koonter, K.D.; Schmitt-Matzen, E.N.; Wagner, A.P.; Holekamp, K.E.; Schmidt, T.M. Symbiotic bacteria appear to mediate hyena social odors. Proc. Natl. Acad. Sci. USA 2013, 110, 19832–19837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Leclaire, S.; Jacob, S.; Greene, L.K.; Dubay, G.R.; Drea, C.M. Social odours covary with bacterial community in the anal secretions of wild meerkats. Sci. Rep. 2017, 7, 3240. [Google Scholar] [CrossRef]
  87. Sankar, R.; Archunan, G. Flehmen response in bull: Role of vaginal mucus and other body fluids of bovine with special reference to estrus. Behav. Process. 2004, 67, 81–86. [Google Scholar] [CrossRef]
  88. Ezenwa, V.O.; Gerardo, N.M.; Inouye, D.W.; Medina, M.; Xavier, J.B. Microbiology. Animal behavior and the microbiome. Science 2012, 338, 198–199. [Google Scholar] [CrossRef]
  89. Verhulst, N.O.; Qiu, Y.T.; Beijleveld, H.; Maliepaard, C.; Knights, D.; Schulz, S.; Berg-Lyons, D.; Lauber, C.L.; Verduijn, W.; Haasnoot, G.W.; et al. Composition of human skin microbiota affects attractiveness to malaria mosquitoes. PLoS ONE 2011, 6, e28991. [Google Scholar] [CrossRef] [Green Version]
  90. Machado, V.S.; Oikonomou, G.; Bicalho, M.L.; Knauer, W.A.; Gilbert, R.; Bicalho, R.C. Investigation of postpartum dairy cows’ uterine microbial diversity using metagenomic pyrosequencing of the 16S rRNA gene. Vet. Microbiol. 2012, 159, 460–469. [Google Scholar] [CrossRef]
  91. Dzięcioł, M.; Niżański, W.; Stańczyk, E.; Kozdrowski, R.; Najder-Kozdrowska, L.; Twardoń, J. The influence of antibiotic treatment of bitches in oestrus on their attractiveness to males during mating. Pol. J. Vet. Sci. 2013, 16, 509–516. [Google Scholar] [CrossRef] [Green Version]
  92. Chenault, J.R.; McAllister, J.F.; Chester, S.T., Jr.; Dame, K.J.; Kausche, F.M.; Robb, E.J. Efficacy of ceftiofur hydrochloride sterile suspension administered parenterally for the treatment of acute postpartum metritis in dairy cows. J. Am. Vet. Med. Assoc. 2004, 224, 1634–1639. [Google Scholar] [CrossRef]
  93. Cepurnieks, G.; Rjabova, J.; Zacs, D.; Bartkevics, V. The development and validation of a rapid method for the determination of antimicrobial agent residues in milk and meat using ultra performance liquid chromatography coupled to quadrupole--Orbitrap mass spectrometry. J. Pharm. Biomed. Anal. 2015, 102, 184–192. [Google Scholar] [CrossRef] [PubMed]
  94. Oliver, S.P.; Murinda, S.E. Antimicrobial resistance of mastitis pathogens. Vet. Clin. North Am. Food Anim. Pract. 2012, 28, 165–185. [Google Scholar] [CrossRef] [PubMed]
  95. Fuller, R. Probiotics in man and animals. J. Appl. Bacteriol. 1989, 66, 365–378. [Google Scholar] [PubMed]
  96. De Seta, F.; Parazzini, F.; De Leo, R.; Banco, R.; Maso, G.P.; De Santo, D.; Sartore, A.; Stabile, G.; Inglese, S.; Tonon, M.; et al. Lactobacillus plantarum P17630 for preventing Candida vaginitis recurrence: A retrospective comparative study. Eur. J. Obstet. Gynecol. Reprod. Biol. 2014, 182, 136–139. [Google Scholar] [CrossRef]
  97. Stapleton, A.E.; Au-Yeung, M.; Hooton, T.M.; Fredricks, D.N.; Roberts, P.L.; Czaja, C.A.; Yarova-Yarovaya, Y.; Fiedler, T.; Cox, M.; Stamm, W.E. Randomized, placebo-controlled phase 2 trial of a Lactobacillus crispatus probiotic given intravaginally for prevention of recurrent urinary tract infection. Clin. Infect. Dis. 2011, 52, 1212–1217. [Google Scholar] [CrossRef]
  98. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [Green Version]
  99. Jembrek, M.J.; Vlainic, J. GABA Receptors: Pharmacological Potential and Pitfalls. Curr. Pharm. Des. 2015, 21, 4943–4959. [Google Scholar] [CrossRef] [Green Version]
  100. Ametaj, B.N.; Iqbal, S.; Selami, F.; Odhiambo, J.F.; Wang, Y.; Gänzle, M.G.; Dunn, S.M.; Zebeli, Q. Intravaginal administration of lactic acid bacteria modulated the incidence of purulent vaginal discharges, plasma haptoglobin concentrations, and milk production in dairy cows. Res. Vet. Sci. 2014, 96, 365–370. [Google Scholar] [CrossRef]
  101. Deng, Q.; Odhiambo, J.F.; Farooq, U.; Lam, T.; Dunn, S.M.; Ametaj, B.N. Intravaginal lactic Acid bacteria modulated local and systemic immune responses and lowered the incidence of uterine infections in periparturient dairy cows. PLoS ONE 2014, 10, e0124167. [Google Scholar] [CrossRef]
  102. Mathys, S.; von Ah, U.; Lacroix, C.; Staub, E.; Mini, R.; Cereghetti, T.; Meile, L. Detection of the pediocin gene pedA in strains from human faeces by real-time PCR and characterization of Pediococcus acidilactici UVA1. BMC Biotechnol. 2007, 7, 55. [Google Scholar] [CrossRef] [Green Version]
  103. Wang, Y.; Ametaj, B.N.; Ambrose, D.J.; Ganzle, M.G. Characterisation of the bacterial microbiota of the vagina of dairy cows and isolation of pediocin-producing Pediococcus acidilactici. BMC Microbiol. 2013, 13, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Esposito, G.; Raffrenato, E.; Lukamba, S.D.; Adnane, M.; Irons, P.C.; Cormican, P.; Tasara, T.; Chapwanya, A. Characterization of metabolic and inflammatory profiles of transition dairy cows fed an energy-restricted diet. J. Anim. Sci. 2020, 98, skz391. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Adnane, M.; Chapwanya, A. Role of Genital Tract Bacteria in Promoting Endometrial Health in Cattle. Microorganisms 2022, 10, 2238. https://doi.org/10.3390/microorganisms10112238

AMA Style

Adnane M, Chapwanya A. Role of Genital Tract Bacteria in Promoting Endometrial Health in Cattle. Microorganisms. 2022; 10(11):2238. https://doi.org/10.3390/microorganisms10112238

Chicago/Turabian Style

Adnane, Mounir, and Aspinas Chapwanya. 2022. "Role of Genital Tract Bacteria in Promoting Endometrial Health in Cattle" Microorganisms 10, no. 11: 2238. https://doi.org/10.3390/microorganisms10112238

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop