Skip to main content

Interactions between reproductive biology and microbiomes in wild animal species

Abstract

Many parts of the animal body harbor microbial communities, known as animal-associated microbiomes, that affect the regulation of physiological functions. Studies in human and animal models have demonstrated that the reproductive biology and such microbiomes also interact. However, this concept is poorly studied in wild animal species and little is known about the implications to fertility, parental/offspring health, and survival in natural habitats. The objective of this review is to (1) specify the interactions between animals’ reproductive biology, including reproductive signaling, pregnancy, and offspring development, and their microbiomes, with an emphasis on wild species and (2) identify important research gaps as well as areas for further studies. While microbiomes present in the reproductive tract play the most direct role, other bodily microbiomes may also contribute to facilitating reproduction. In fish, amphibians, reptiles, birds, and mammals, endogenous processes related to the host physiology and behavior (visual and olfactory reproductive signals, copulation) can both influence and be influenced by the structure and function of microbial communities. In addition, exposures to maternal microbiomes in mammals (through vagina, skin, and milk) shape the offspring microbiomes, which, in turn, affects health later in life. Importantly, for all wild animal species, host-associated microbiomes are also influenced by environmental variations. There is still limited literature on wild animals compared to the large body of research on model species and humans. However, the few studies in wild species clearly highlight the necessity of increased research in rare and endangered animals to optimize conservation efforts in situ and ex situ. Thus, the link between microbiomes and reproduction is an emerging and critical component in wild animal conservation.

Introduction

Animal evolution has taken place in environments replete with microorganisms. With a few exceptions (in some insects, for example), every animal species is colonized and lives in close association with microbial partners that include viruses, bacteria and microscopic eukaryotes [1]. Animal biology cannot be understood without reference to the microbial communities that have evolved to live on and within hosts, usually referred to as the organism’s microbiomes. This has led researchers to propose that the amalgam of host and the microbial community genomes might be a unit of selection [2]. The term microbiome is generally used to refer to both the composition of a microbial community (also termed the microbiota) and the entire microbial genomic content of the community (also termed the metagenome). The expressed microbial genome (also termed the metatranscriptome) is the fundamental biological unit of importance but is more difficult and expensive to determine compared to identifying the microbiota by 16S rRNA gene sequencing. The present review will be using the term ‘microbiome’ to refer to the microbial community composition.

The microbiome is a valuable proxy for estimating physiological states, including in reproduction, which is essential to the survival of species. Balanced interactions within and between host cells and non-host cells, such as the resident microbial community, are essential to overall fitness, including reproductive health. Reproductive microbiomes are defined as microbial communities inhabiting host reproductive tracts in males and females [3, 4]. However, other microbial communities (e.g., gut, milk, and glandular microbiomes) can further influence reproductive success from mate choice to healthy pregnancy and successfully producing offspring [2]. For example, signal-producing microbiomes (in e.g., scent glands) may play a role in pre-copulatory reproductive communication. Similarly, the milk microbiome influences post-natal infant development. Host-associated microbiomes, including reproductive microbiomes, are also affected by both endogenous processes, such as host physiology [5, 6] and exogenous factors, such as social interactions and environmental variations [7, 8]. Thus, while microbiomes present in the reproductive tract may play the most direct role, other bodily microbiomes may contribute to facilitating reproduction. Figure 1 illustrates the complex interactions between host microbiomes, life cycles, and reproductive success. It is recommended that research on reproductive microbiomes should also be expanded to include microbial effects before copulation/fertilization and the transfer of appropriate microbiomes to the offspring.

Fig. 1
figure 1

interactions between host microbiomes, life cycles, and reproductive success in wild animal species

Some of the earliest observations on the linkages between host microbiota and host reproduction occur in freshwater hydra, in which bacteria are essential to the production of offspring through asexual budding [9]. Since then, a growing body of literature on the role of microbiota in host reproduction has been produced, with a large focus on invertebrate systems (particularly Wolbachia symbioses) and vertebrate model organisms [10]. This is in part due to the ability to perform controlled laboratory experiments in these systems that allow for inference regarding host reproductive-microbiota interactions. Not surprisingly, we know little about microbiomes in animal species compared to human microbiomes. Most reports on the reproductive effects of microbiomes of non-human species are on domesticated and laboratory animals, mostly mammals [3].

Although the number of microbiome studies conducted on wildlife remains lower than in other animal species, it has increased over the last decade [3]. Pronounced animal species declines and loss of natural habitats across the globe have led to the establishment of many captive assurance populations in which reproductive success is critical to the persistence of rare and endangered vertebrate animals [11]. Captive breeding is known to alter host-associated communities and there is increasing support for an associated effect on host reproductive success [12]. As observed in humans [13, 14], assisted reproductive technologies (from hormonal stimulation to embryo transfer), contraceptive methods, and treatment of pathologies may have an impact on the microbiomes. Our attempts to breed species in captivity (naturally or through assisted reproductive technologies) may have significant effects on the species microbiomes that will influence success or failure of reproduction.

The objective of this review is to (1) specify the interactions between animals’ reproductive biology, including reproductive signaling, pregnancy, and offspring development, and their microbiomes, with an emphasis on wild species and (2) identify important research gaps as well as areas for further studies. We will begin by discussing the links between microbiomes, life stage, and reproductive status in males and females. Then, we will move to detailing the link between microbiomes, reproduction, and environmental changes. We will end by discussing future directions in that area. Throughout the manuscript we also iterate the importance of including appropriate environmental controls and experimental criteria to improve the validity of reproductive microbiome research, since these are typically low biomass [15, 16].

Links between microbiomes, life stage, and reproductive status in males and females

Endocrine mediation of reproductive microbiomes across reproductive cycles or breeding seasons

The host endocrine system affects the structure of reproductive microbiomes in many animal species [5, 6], particularly through corticosteroid hormones (glucocorticoids and mineralocorticoids) [17, 18]. Glucocorticoids, such as cortisol and corticosterone, are associated with stress and can affect reproductive functions through known links to reproductive behavior and endocrine cycling as seen in primates, rhinos, and wild birds [6, 19,20,21,22,23]. [8]Evidence for bidirectional signaling between the microbiome and the hypothalamic pituitary axis and greater gut-brain axis also suggests that microbiomes can have significant effects on the host endocrine system [6]. Glucocorticoid-related microbiome effects have largely been studied in the gut microbiome with some investigation of reproductive outcomes in rhinos [23], fish [24], birds [22, 25], and pigs [26]. For example, in Eastern black rhinos (Diceros bicornis michaeli), glucocorticoid and progestagen concentrations, as well as the abundance of certain less common gut microbes, differ between female rhinos who produce calves and females that are considered non-breeders [23]. This suggests a relationship between hormone cycling, reproduction, and gut microbiome; however, the direction of these effects is unknown [23]. Bacteria can metabolize steroid hormones such as glucocorticoids and convert them into androgen hormones in humans [27], highlighting the challenge of disentangling hormone origin and the cause-and-effect relationships between microbiomes and reproduction.

Studies in humans also indicate that increased cortisol associated with prolonged stress can influence the vaginal mucosa, including the deposition of glycogen, which mediates the abundance of Lactobacillus spp. and underpins aspects of human vaginal tract health and copulatory success [28]. More broadly, increased cortisol resulting from prolonged stress is correlated with changes in gut microbiome structure and inflammatory responses, which, in turn, are known to affect pregnancy outcomes in women [29]. Although more research is needed in wild animal species, the relationships between glucocorticoid hormones and microbiomes provide plausible mechanisms by which psycho-social and physiological stress can influence health and reproductive outcomes.

Likewise, sex hormones play an integral part in structuring reproductive microbiomes. Previous research has largely been focused on female mammalian microbiomes and their relationships with ovarian hormones (estradiol and progesterone), reproductive cyclicity, or general variation in vaginal microbiomes, such as in non-human primates [7] and giant pandas (Ailuropoda melanoleuca) [30]. In primates, the increased circulating estradiol associated with sexual maturity in female mammals is linked to changes in vaginal epithelium function (e.g., increased glycogen production) and microbial community structure, further highlighting the complex relationships between endocrine function, host physiology, and microbial communities [7]. In mice models, wild-type and estrogen receptor knock-out mice even indicate an effect of estrogen binding on microbiome structure [31].

In males, testosterone concentrations are linked to seminal microbiome structure, sperm characteristics (see below), and infertility [32,33,34]. However, those studies still need to be confirmed with appropriate controls to discount the influence of DNA contamination on the results as seen in research on other microbiomes. In male birds, cloacal microbial diversity is positively correlated with testosterone levels, which in turn may affect rates of reproductive behaviors—such as extra-pair copulations—that increase opportunities for microbial transmission [35]. In free-living rufous-collared sparrows (Zonotrichia capensis), cloacal bacterial communities differ between the sexes when they are in breeding condition. In males, but not in females, the bacterial community becomes more diverse with the onset of reproduction, and then decreases in diversity as males transition to non-breeding condition [36].

Interactions between sex hormones and microbes can have significant metabolic effects on the host. In female Phayre’s leaf monkeys (Trachypithecus phayrei) reproductive hormones, specifically progestagens, contribute to the shifts in the gut microbiome during pregnancy and lactation [37]. There also is a shift in the maternal gut microbiome of Tibetan antelope (Pantholops hodgsonii) during the perinatal period [38], though the effects on reproduction are unknown. Future research over the next decade will likely elucidate how host endocrine-microbiome interactions affect host physiology and reproduction. At present, evidence for sex hormone-microbiome relationships in both sexes largely stem from studies of humans, whose reproductive microbiomes differ drastically even from other closely related primates (e.g., the unique Lactobacillus dominance in many, but not all, human vaginal microbiomes; [39]). The extent to which these patterns of endocrine-microbiome relationships apply to endangered species warrants further investigation, especially given the common use of husbandry practices that alter endocrine function (e.g., hormonal contraceptives; see below).

Microbiomes and reproductive signaling (visual and olfactory)

Bodily microbiomes could potentially contribute to reproduction by mediating pre-mating reproductive signaling. Bodily microbiomes could potentially contribute to reproduction by mediating pre-mating reproductive signaling (e.g., plumage or bill color in birds [2, 40]). The strongest examples for reproductive signaling mediated by the microbiome stems from studies on olfaction. Olfactory signaling is a mode of reproductive communication that has long been thought to rely on a combination of volatile organic compounds produced by the host and by their commensal microbes (i.e., the fermentation hypothesis; [41]). Olfactory chemical cues can facilitate mate choice and reproductive success through ‘honest’ signaling of animal health, kinship and genetic compatibility, social status, and reproductive cycle and fertility [42,43,44]. Depending on the social organization of the animal species, these cues are relevant to and can be produced by one or both sexes. Although much of the previous research has been focused on male odor production to attract female mates, females of many species (including primates that are female-dominant) also rely on scent signaling to convey socially relevant information [45]. These cues can emanate from multiple body sites (e.g., scent gland and skin surfaces) and waste products (e.g., urine and feces) [46]. Scent glands, in particular, provide protected, ideal habitats for microbial growth and specialized glandular tissue can occur at various body sites, including homologous sites in males and females (e.g., labial glands in females and scrotal glands in males).

In multiple mammals, scent gland signals covary with glandular bacterial communities [47, 48]. These olfactory signals convey social and reproductive information that may influence social status and mate choice (for example in giant pandas [49] or European badgers [50]). In male diademed sifakas (Propithecus diadema) sternal gland microbiomes and olfactory chemical signals vary according to the male’s breeding vs. non-breeding status [51]. Similarly, in Chinese musk deer (Moschus berezovskii), musk gland bacteria and musk chemical composition reflected breeding status as mated vs. unmated, an important signal for mate choice [52]. Thus, scent gland microbes are associated with the host’s ability to advertise reproductive information, which may affect their reproductive success. Outside of mammalian species, bacteria from the uropygial glands of multiple bird species produce volatile compounds that are strongly linked to reproductive success [53] and appear to correlate with major histocompatibility complex diversity and reproductive compatibility [54]. Additionally, in a common South American tree frog (Dendrobates sp.), sex-specific scents can be traced to a frog-specific skin bacterium that produces a class of compounds with strong odor properties [55]. However, investigations of microbial contributions to scent signaling are scarce, particularly in wild animals. If, as mounting evidence suggests, host-associated microbes underpin signals of reproductive status and mate choice, investigation of this microbe-olfaction-reproduction axis may be especially relevant to holistic perspectives of reproductive success.

Influence of mating behavior and copulation on microbiomes

In female mammals, there is a complex vaginal microbiome, which is vital to many aspects of reproduction, including preparing the host for reproductive activity. The disruption of the vaginal microbial community can have severe consequences for health and reproductive success [56,57,58]. The vaginal microbiome has been studied in a number domesticated mammal species, non-human primates and the giant panda (reviewed in [59]). Vaginal microbiomes vary widely among host taxa, and closely related species will not necessarily have similar vaginal microbiomes [60]. A Lactobacillus dominated vaginal microbiome appears unique to humans [7, 60] and even within humans there are stable, healthy vaginal microbiomes with few Lactobacillus [61].

In male mammals, semen is a complex biological fluid that contains nutrients for the spermatozoa (fructose, glucose, amino acids, minerals) factors that affect sperm motility and potency, and has a characteristic pH [62,63,64,65]. Many of the characteristics that make semen an adaptive medium for sperm cells also are conducive to microbial organisms. Certainly, pathogens responsible for many sexually transmitted infections (STIs) live in seminal fluid. The existence of microbes in seminal fluid implies that a microbiome exists in the seminal vesicles and/or other parts of the male urogenital tract [31]. Most of the research on seminal fluid microbiomes has been done in humans and rodent biomedical models, and showed microbial-associated sperm dysfunction [2]. Evidence from mice suggests that the seminal fluid microbiome may also have beneficial effects on reproduction [31]. Studies in men have found associations between microbial taxa and sperm concentration, motility, and quality. Some microbes are associated with a reduction in sperm quality, whereas others are associated with higher quality sperm, suggesting that certain microbes may be indicators of sperm quality [32, 66]. A recent study conducted in peccaries (Pecari tajacu) also highlighted the relation between preputial microbiome and semen quality [67]. As mentioned earlier, more studies are warranted to discount the influence of DNA contamination on the results (as seen in research on other microbiomes). Furthermore, the seminal microbiome may also have long term effects on fetal development that may influence later health and reproductive success of offspring [31]. Male mice fed a high-fat diet produced male offspring with poorer sperm quality [68], suggesting that host metabolism has effects on semen and its microbiome [31]. It is unknown whether these two effects are causally related, but it raises the possibility that the seminal microbiome may have next generational effects on reproductive success.

Copulation is a necessary component of vertebrate reproduction and also presents an opportunity for the transmission and interactions of microbes between host males and females in both directions. The transfer of microbes by copulation—and the associated risk of perturbing the established microbiome or STIs—likely exerts selective pressures on male and female reproductive microbiomes [7, 39]. In non-human primates, peak sexual receptivity and copulation behavior coincide with increased vaginal microbial diversity [69, 70], which may defend against the increased risk of microbial transmission via copulation, including STIs [71]. In owl monkeys (Aotus sp.), unmated males and females differ from mated animals in preputial and vaginal microbiomes, respectively, which was a result of the transfer of microbes via copulation [72]. Whether the microbiome within semen can benefit the female partner is unknown. Although sexual microbial transfer is most widely studied from the perspective of STIs, copulation may also be an important vector for the transfer of benign or even beneficial microbes [56]. Certainly, the microbial community within seminal fluid will at least briefly inhabit the female reproductive tract after copulation and may potentially influence fertilization and perhaps even implantation. Evidence from human studies suggests that the seminal and vaginal microbiomes of consistent sexual partners exhibit similarities and that the vaginal microbiome changes slightly after intercourse [73]. More numerous or frequent copulations (as seen in promiscuous species) have been linked to greater vaginal or cloacal microbial diversity (in mice: [74]; in primates [60, 75]; in lizards [76]). Greater vaginal microbiome diversity in promiscuous species can aid in creating a community that is more resilient to perturbation or infection and, ultimately, may improve reproductive success [71]. However, in humans, promiscuity has been correlated with dysbiosis of vaginal microbiomes [77], suggesting that frequent copulations with different partners could also disrupt or destabilize the vaginal microbial community.

Pregnancy and birth are major colonizing events of microbiomes

The lower mammalian female reproductive tract (vulva, vagina and cervix) harbors complex, dynamic microbiomes which vary by region, hormonal state, and reproductive condition [58, 78]. A lack of stability in the vaginal microbiome in early pregnancy, especially with any significant presence of pathogenic strains, is associated with pregnancy complications including preterm birth and spontaneous abortion [79]. In contrast to the lower tract, the upper mammalian female reproductive tract (uterus, fallopian tubes) is considered a protected body region, with the expectation that it will be difficult for microbes to travel from the lower into the upper reproductive tract and become established. This does not mean that microbes cannot migrate along the tract or colonize these regions, but the diversity and density of the microbes in the upper reproductive tract is generally sparse compared to other body regions in women [80]. The developing fetus, while not in a sterile environment, is in a protected environment, and thus exposed to a lower abundance and diversity of microbes.

There is consistent evidence for a uterine wall microbiome in several species, including humans, horses, cows and rhesus macaques (Macaca mulatta) [59, 81]. The uterine microbiome is distinct from and is less densely populated than the vaginal microbiome [59]. There is evidence in both humans and horses for an association between uterine microbiome structure and successful pregnancy outcome [59]. However, more research is required to investigate the robustness of and mechanisms underlying this association.

There is controversy regarding the extent and stability of the microbial communities found in the mammalian placenta. Note that most of the evidence for or against a placental/fetal microbiome again comes from human data. A placental microbial community has consistently been found in human pregnancies that are complicated by pre-eclampsia and preterm birth (reviewed in [82]) and microbial invasion of the placenta is considered a causal aspect for these pathologies; however, several studies also reported the existence of a placental microbiome during uncomplicated pregnancy [83, 84]. Studies also found evidence of microbial communities in amniotic fluid [85]. However, subsequent studies have challenged these results, finding no detectable microbiota in amniotic fluid from uncomplicated pregnancies with intact membranes [86, 87]. A recent systematic review concluded there was no evidence for a placental microbiome in uncomplicated human pregnancy [82]. While microbial communities may exist in the uterus, the placenta, and the amniotic sac, these sites are largely sterile in humans [82, 88]. A study of uterine, placental and fetal tissue from four rhesus macaque pregnancies delivered by Cesarean section at approximately 130 days of the normal 166-day gestation found a uterine wall microbiome, but no evidence of placental or fetal microbial communities [81]. Recent studies in horses found evidence for microbiomes in placenta, amniotic fluid and meconium, indicating early establishment of a microbiome within the fetal gut by microbes of placental origin (reviewed in [89]). However, these studies were done on material collected after parturition, the abundance of microbes was low and thus the possibility of contamination cannot be discounted. There is no evidence for any placental microbiome in cattle [90] and bacterial loads of amniotic fluid were no different from negative controls in calves delivered by Cesarean section [91]. However, there are detectable, albeit low abundance, microbes in calf meconium [91]. Thus, colonization of the placenta, amniotic sac, and fetus by maternal microbes may reflect pathology rather than adaptive function. An ascending intrauterine infection is a risk factor for preterm birth and is associated with both maternal and fetal inflammatory response [92]. Growing evidence suggests that the interactions between maternal reproductive tract microbiomes and maternal and fetal immune response are important factors in birth outcome [92]. Prenatal exposure to microbes happens in pregnancies complicated with certain pathologies, but whether microbial communities in placental tissue can inoculate the mammalian fetus before birth remains uncertain.

Regardless of the mode of transmission, early life microbiome may impact later reproduction, as shown in the mouse model [93]. The microbes acquired at birth can affect offspring health later in life (as reviewed by [94]), and health status can have subsequent effects on reproductive success, such as obesity [95]. When the fetus travels through the reproductive tract it will become inoculated with the maternal microbiome [96]. In humans, infants born vaginally vs. by Cesarean section will have different skin and gut microbiomes [97, 98]. For egg laying vertebrates there is less evidence, but both lizard and bird eggs appear to harbor bacterial communities inside the shell [99]. Presumably this inoculation occurs in the upper regions of the oviduct before the shell has formed. The extent to which any maternal microbes transferred after shell formation can persist in the nest and thus be available to inoculate hatchlings is unknown. Similarly, Bornean foam-nest breeding frogs deposit eggs within foam nests with the internal nest colonized by bacteria (McGrath et al. in review for this special issue). The vertical transmission of the maternal microbiome to offspring is a critical aspect of successful reproduction, and the microbiome of the female reproductive tract is the first exposure for eggs and fetuses.

Post-partum microbiomes and infant development/reproductive success, from mother to child

In contrast with the often brief exposure to maternal vaginal microbiomes, infant exposure to the maternal skin and oral microbiomes will be chronic, occurring over the much longer time period of neonatal dependence, but also within the context of well-developed (though still developing) neonatal microbiomes that may be capable of resisting colonization by novel microbes. Maternal skin contact may be more important for certain taxa. For example, in southern hairy-nosed wombats (Lasiorhinus latifrons), pouches contain communities of microorganisms that are substantially altered by the host reproductive cycle. More investigations into the roles that pouch microorganisms may play in marsupial reproductive health and joey survival are warranted [100].

The neonate will also be exposed to a microbiome unique to mammals, the milk microbiome. Milk contains a microbial community that can vary among mammalian species [101]. The origin of the milk microbiome is not well established, but evidence from rodent models suggests that bacteria may be transported from the maternal gut to the mammary gland [102]. Compared to the vaginal microbiome the milk microbiome is depauperate, both in terms of species numbers and total amount of microbial cells. However, a breastfed baby is estimated to consume 105 to 107 bacteria each day [103]. Suggested biological functions for the milk microbiome include inoculation of the neonatal gut, activation of mammary immune systems and cells [58], and stimulation of the developing neonatal immune system [104]. Another potential function is to develop immune tolerance in the neonate to commensal and symbiotic maternal microbes. Note that the milk microbiome is not the only aspect of milk that influences the neonatal gut microbiome. Milk contains live maternal immune cells, maternal immune function molecules, antibiotic molecule such as lysozyme, and prebiotic oligosaccharides that are not metabolized by the neonate but rather by the neonate’s gut microbes [58]. The relative importance of the milk microbiome compared to these other aspects of milk are not known. The transfer of a gut microbiome from parent to offspring through feeding is not just a mammalian trait. Several avian taxa regurgitate food into their chicks and produce crop secretions, termed ‘crop milk’ that they feed to chicks. These secretions contain similar bioactive constituents to mammal milk and contain microbes [105, 106]. All these exposures to maternal microbiomes shape the offspring microbiomes, which, in turn, affects later health.

There also are extensive interactions between microbes, their products and the host immune system that are often symbiotic, not antagonistic. The lack of a microbiome can disrupt neonatal development and gnotobiotic rodent models display significant deficiencies in anatomy, physiology, metabolism and immune function [107]. The development of the neonatal immune system appears to be highly influenced by microbiome effects [107]. Captive breeding programs for endangered species need to consider how management procedures may affect early life microbiomes.

Link between microbiomes, reproduction, and environmental changes

Captivity, microbiomes, and fertility control

Often attributed to the loss of native or beneficial microbes, the effects of captivity vary across host species but are undeniably a vital aspect of conservation breeding [108]. Moreover, efforts to manage or improve reproduction in captive populations, via contraceptives or assisted reproduction, can shape reproductive microbiomes with largely unknown consequences [3]. Beyond the scope of captivity, human activity, including the alteration and contamination of natural environments, can influence animal microbiomes and affect reproductive success; however, empirical links between these anthropogenic influences and reproductive microbiomes are currently sparse. In the face of extensive and continuing human influence on natural ecosystems and their inhabitants, there has been a recent and urgent call for greater ‘microbiome conservation’ in order to characterize and preserve global microbial diversity [99, 109, 110]. Here, we reinforce this call and suggest that the study and conservation of reproductive microbiomes are particularly relevant to endangered or rare species in which managed breeding programs are vital resources for species survival.

Understanding the effects of captivity on host-associated microbiomes is crucial to maintaining and improving the care and management of endangered species [108]. The majority of research to date has focused on gut microbial communities [108, 111] with few studies examining captivity-induced changes in reproductive microbiomes [112]. Nevertheless, burgeoning evidence of ‘cross-talk’ between gut and reproductive microbiomes suggests that many gut-centered outcomes of captivity can have downstream effects on the suite of reproductive microbiomes mentioned above [113, 114]. The factors driving gut microbiome variation between captive and wild animals (e.g. dietary mismatches; [115]) may similarly influence reproductive microbiomes by altering host metabolism and dictating available resources. For instance, Williams et al. [12] found that in southern white rhinoceros (Ceratotherium simum simum) fertility varied significantly with phytoestrogen profiles, the abundance of several gut bacterial taxa, and microbially-derived phytoestrogen metabolites. This suggests that diet may drive transformation of dietary phytoestrogens by the gut microbiome which then impacts reproductive outcomes. Even pre-copulation, if the effects of captivity alter scent gland microbial communities similarly to the widely reported effects seen in the gut microbiomes, associated olfactory signals could be altered, resulting in discordant signals and a disruption of reproductive communication.

Medical treatments received by animals in captivity also have the potential to shape the structure and function of reproductive microbiomes [70, 108]. Antibiotic treatment has been strongly linked to dysbiotic microbial communities, and thus antibiotic use in captive endangered animals may structure reproductive microbiomes and influence associated outcomes. For example, antibiotic treatment in captive primates result in alterations of gut microbiomes, with potential long-term consequences for gut health [116]. There also is evidence in lactating women that prenatal antibiotic use changes the milk microbiome, by decreasing the prevalence of beneficial microbes and increasing the proportion of potential pathogens [117]. Although very little is known about the prevalence and impact of antibiotic resistance genes in reproductive microbiomes, they may pose a particular risk to endangered animals in captivity, where the efficacy of antibiotic treatment is paramount to successful care and husbandry.

Additionally, artificial fertility control, via e.g., contraceptives, may further influence the reproductive microbiomes of captive animals. Hormonal contraceptives supply artificial reproductive hormones (e.g., depot medroxyprogesterone acetate) that disrupt natural endocrine processes to minimize fertility and prevent pregnancy. In hormonally contracepted humans, endocrine mediation of microbial communities is altered resulting in changes in the composition of reproductive microbiomes [118, 119]; however, evidence for the effects of contraceptives on the microbiomes of non-human animals is scarce. In humans and non-human primates, hormonal contraception has been linked to the dampening or disruption of olfactory cues used for reproductive communication [120,121,122] potentially reflecting changes in odor-producing bacteria. Given the propensity for reproductive dysfunction in many captive, endangered species, greater study of reproductive microbiomes in these settings can shed light on potential mechanisms of dysbiosis and, in turn, provide opportunities to integrate microbial ecology into successful husbandry practices.

Environmental contaminants, microbiomes, and reproductive health

Environmental contaminants, including herbicides, pesticides, and heavy metal pollutants, can cause adverse effects on reproduction in wildlife [123, 124] and change host microbiomes [125,126,127]. The adverse effects on reproduction may be mediated or modulated by changes in the host microbiome [128]. For instance, gut microbes can metabolize environmental chemicals and thus modulate toxicity for the host animal [129]. Likewise, one of the most commonly used herbicides, glyphosate, inhibits the biochemical pathway for aromatic acid synthesis which is found in numerous bacterial species and can subsequently alter host-associated microbiomes [126]. Overall, the interactions among reproduction, contaminants and microbiomes have rarely been examined. One study found that glyphosate-based herbicides altered gut microbiomes and reproductive hormone levels in Japanese quails (Coturnix japonica) in a long-term study, but did not directly affect reproduction in either sex (maturation, testis size or egg production) [130]. Future studies on the effects of environmental contaminants on reproductive microbiomes should consider that these interactions may be subtle or potentially delayed in order to fully understand the risks related to contaminant exposure [131].

In addition, man-made antibiotics that leach into natural ecosystems can alter both environmental and host-associated microbial communities and facilitate the spread of antibiotic resistance genes [132, 133]. In contrast to the effects of antibiotic treatment done under veterinary care, antibiotic contamination of the environment can exert selective, but often non-lethal pressure on microbes, allowing them to evolve or spread resistance genes and expand the ‘resistomes’ in natural environments. It is now well-known that antibiotic-resistant bacteria in environmental settings (e.g., water, soil, food items) can be picked up by animals and humans [134,135,136], where they can be incorporated into host-associated microbiomes and potentially pose significant health risks [133, 137]. Indeed, in wild and captive populations of endangered ring-tailed lemurs (Lemur catta), antibiotic resistance genes covaried between host gut microbiomes and environmental soil microbiomes, a pattern that also reflected anthropogenic use of antibiotics and environmental contamination [138].

Pathogens, microbiomes, and reproductive health

There are well known proximate mechanisms by which pathogens can influence reproduction, usually negatively. When animals mount an immune response due to pathogenic infection, animals can increase, and maintain or decrease their reproductive investment. Such life-history tradeoffs in energy allocation between reproduction and fighting infection are fundamental to fitness. We can broadly consider pathogens as part of the host microbiome as they often colonize and live as part of the microbial community during infection. Most studies to date on reproduction-infection tradeoffs have examined this in the context of single pathogen infections or perceived infection (e.g., vaccination). Evidence shows that trade-offs between reproduction and fighting infection go in all directions, and likely reflect unique pressures, such as age of individuals in male crickets [139].

A commonly tested hypothesis is that an individual will increase reproductive investment when immuno-compromised [140]. The terminal investment hypothesis predicts that individuals should invest more in their present reproduction if they are less likely to survive, with infection and disease being a potential cue of diminishing reproductive value. A variety of studies provide support for the terminal investment hypothesis in reproductive efforts, including in frog species infected with a fungal pathogen that up-regulate gametogenesis [141], increase male calling behavior as well as mating success [142]. Likewise, similar support exists in bird species. For example, great tits (Parus major) infected with haemosporidian parasites have increased reproductive success [143], blue footed boobies (Sula nebouxii) with elicited immune response increase parental care [144], and vaccinated house sparrows (Passer domesticus) increased egg laying efforts [145]. On the other hand, mounting an immune response can lead to reduction in reproductive investment. Another intriguing hypothesis is the immuno-competence handicap hypothesis which predicts that only high-quality males can maintain high levels of testosterone and afford the physiological cost of hormone-derived immunosuppression, but similar to the terminal investment hypothesis data remains ambiguous to support immunocompetence handicap hypothesis [146]. Future work examining how the larger collective microbial community is involved in these tradeoffs would be useful.

Conclusions—future directions

While microbiomes present in the reproductive tract play the most direct role, other bodily microbiomes may contribute to facilitating reproduction in wild animal species. Endogenous processes related to host physiology can both influence and be influenced by the structure and function of microbial communities. Importantly, exposures to maternal microbiomes (through the vagina, skin, and milk) shape the offspring microbiomes later in life (Fig. 1). In many cases, we do not know if shifts in the microbiome are the cause of changes in the host reproductive status or a by-product of physiological changes.

In addition, host-associated microbiomes are also influenced in wildlife by exogenous factors, such as social interactions, environmental variations, or ex situ breeding programs. However, much more research is warranted in that area in order to optimize conservation efforts. Although our review is biased towards mammals, as there currently is more information regarding reproductive microbiomes, many of the concepts discussed in this review (as well as the next steps proposed below) could be widely applied to other vertebrates.

It is important to remember that there is still a need for more comprehensive microbe taxonomy (for better identification) and more studies about microbial functions. It is critical to culture as many of these ‘reproductively relevant’ microbes as possible and use standardized 16S protocols so we can compare across studies and find shared microbes. As mentioned earlier, it also is essential to reduce contamination bias, including appropriate environmental controls and experimental criteria to improve the validity of low microbial biomass research [15, 16]. We also want to further highlight the importance of using adequate controls in sampling and sequencing reproductive microbiomes. To improve ‘credibility’ of reproductive microbiomes – (1) numerous negative control samples should be taken throughout sampling and throughout procedures in the laboratory to be used to identify potential contaminants post sequencing, such as by using the package ‘decontam’ available in R [147] and (2) use of positive mock bacterial community controls to verify distribution of communities are not overly skewed and to verify spurious sequences are not being generated during molecular procedures in preparing libraries for sequencing.

To fill the gaps of knowledge in wildlife reproduction and microbiomes, we propose a list of topics that should be explored to better understand and possibly manipulate microbiomes in wild animal species:

  • Relationship between hormones, reproduction, and gut microbiome in males and females (including the impact of hormonal contraception or stimulations).

  • Effect of puberty and aging on microbiomes [148, 149].

  • Microbial contributions to scent signaling and reproduction.

  • Role of male and female microbiomes on sperm competition during and after mating.

  • Role of seminal microbiome on the reproductive success of offspring (transgenerational effect).

  • Role of microbiome during conception, early embryo development in the oviduct, implantation, delayed implantation [150].

  • Association between uterine microbiome structure and successful pregnancy outcomes (leading to the development of non-invasive microbial markers of early pregnancy and fetal health, prediction of infant health).

  • Influence of maternal diet on maternal and fetal microbiomes during pregnancy.

  • Extent and stability of the microbial communities found in the mammalian placenta.

  • Investigations into the function of the pouch microbiomes in marsupials.

  • Exploration of the amphibian cloacal and spermic urine microbiome.

  • Microbiomes and reproduction in invertebrates (e.g., oysters, corals).

  • Consequences of captive breeding management on early life microbiomes and future reproductive fitness.

  • Prevalence and impact of antibiotic resistance genes in reproductive microbiomes and their potential risk to endangered animals in captivity).

  • Links between infertility or reproductive issues and microbiomes.

  • Impact of artificial insemination or embryo transfer on microbiomes during gestation (does artificial insemination provide the same microbial signals as copulation?). Effects of semen microbiomes on in vitro fertilization.

  • Manipulating or bioengineering the microbiomes to enhance or suppress fertility [151].

Lastly, in situ conservation efforts could benefit from more studies on interactions between reproduction, contaminants, and microbiomes. This could be conducted on sentinel species that could reflect the status of reproductive health in wild species sharing the same habitat. Application of ecological theory and advancement of analytical tools would help to understand what is structuring microbiomes [152, 153]. Collective findings will inform strategies to improve reproductive health in conservation breeding or in the wild. It will also help to develop conservation policy or legislation to include microbiome assessments for translocation or reintroduction of wild animal species in their natural habitats.

Availability of data and materials

Not applicable.

References

  1. Hammer TJ, Sanders JG, Fierer N. Not all animals need a microbiome. FEMS Microbiol Lett 2019; 366.

  2. Rowe M, Veerus L, Trosvik P, Buckling A, Pizzari T. The reproductive microbiome: an emerging driver of sexual selection, sexual conflict, mating systems, and reproductive isolation. Trends Ecol Evol. 2020;35:220–34.

    PubMed  Google Scholar 

  3. Comizzoli P, Power M. Reproductive microbiomes in wild animal species: a new dimension in conservation biology. Adv Exp Med Biol. 2019;1200:225–40. https://doi.org/10.1007/978-3-030-23633-5_8.

    Article  PubMed  Google Scholar 

  4. Berg G, Rybakova D, Fischer D, Cernava T, Vergès MCC, Charles T, Chen X, Cocolin L, Eversole K, Corral GH, Kazou M, Kinkel L, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:1–22.

    Google Scholar 

  5. Neuman H, Debelius JW, Knight R, Koren O. Microbial endocrinology: The interplay between the microbiota and the endocrine system. FEMS Microbiol Rev. 2015;39:509–21.

    PubMed  Google Scholar 

  6. Williams CL, Garcia-Reyero N, Martyniuk CJ, Tubbs CW, Bisesi JH. Regulation of endocrine systems by the microbiome: Perspectives from comparative animal models. Gen Comp Endocrinol 2020; 292.

  7. Stumpf RM, Wilson BA, Rivera A, Yildirim S, Yeoman CJ, Polk JD, White BA, Leigh SR. The primate vaginal microbiome: comparative context and implications for human health and disease. Am J Phys Anthropol. 2013;152:119–34.

    PubMed  Google Scholar 

  8. Stothart MR, Palme R, Newman AEM. It’s what’s on the inside that counts: Stress physiology and the bacterial microbiome of a wild urban mammal. Proceedings of the Royal Society B: Biological Sciences, vol. 286. Royal Society Publishing; 2019.

  9. Rahat M, Dimentman C. Cultivation of bacteria-free Hydra viridis: missing budding factor in nonsymbiotic Hydra. Science (80-). 1982;216:67–8.

    CAS  Google Scholar 

  10. Stouthamer R, Breeuwer JAJ, Hurst GDD. Wolbachia pipientis: Microbial manipulator of arthropod reproduction. Annu Rev Microbiol. 1999;53:71–102.

    CAS  PubMed  Google Scholar 

  11. Comizzoli P, Brown JL, Holt WV. From the ivory tower to reality! Conclusions of the new edition. Adv Exp Med Biol. 2019;1200:545–550. https://doi.org/10.1007/978-3-030-23633-5_18.

  12. Williams CL, Ybarra AR, Meredith AN, Durrant BS, Tubbs CW. Gut Microbiota and phytoestrogen-associated infertility in southern white rhinoceros. MBio 2019; 10.

  13. Sirota I, Zarek SM, Segars JH. Potential influence of the microbiome on infertility and assisted reproductive technology. Semin Reprod Med. 2014;32:35–42.

    PubMed  PubMed Central  Google Scholar 

  14. Franasiak JM, Werner MD, Juneau CR, Tao X, Landis J, Zhan Y, Treff NR, Scott RT. Endometrial microbiome at the time of embryo transfer: next-generation sequencing of the 16S ribosomal subunit. J Assist Reprod Genet. 2016;33:129–36.

    CAS  PubMed  Google Scholar 

  15. Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, Turner P, Parkhill J, Loman NJ, Walker AW. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014;12:1–12.

    Google Scholar 

  16. Eisenhofer R, Minich JJ, Marotz C, Cooper A, Knight R, Weyrich LS. Contamination in low microbial biomass microbiome studies: issues and recommendations. Trends Microbiol. 2019;27:105–17.

    CAS  PubMed  Google Scholar 

  17. Brotman RM, Ravel J, Bavoil PM, Gravitt PE, Ghanem KG. Microbiome, sex hormones, and immune responses in the reproductive tract: Challenges for vaccine development against sexually transmitted infections. Vaccine. 2014;32:1543–52.

    CAS  PubMed  Google Scholar 

  18. Sandrini S, Aldriwesh M, Alruways M, Freestone P. Microbial endocrinology: Host-bacteria communication within the gut microbiome. J Endocrinol. 2015;225:R21–34.

    CAS  PubMed  Google Scholar 

  19. Weingrill T, Gray DA, Barrett L, Henzi SP. Fecal cortisol levels in free-ranging female chacma baboons: relationship to dominance, reproductive state and environmental factors. Horm Behav. 2004;45:259–69.

    CAS  PubMed  Google Scholar 

  20. Arlet ME, Grote MN, Molleman F, Isbell LA, Carey JR. Author’s personal copy Reproductive tactics influence cortisol levels in individual male gray-cheeked mangabeys (Lophocebus albigena) 2008.

  21. Grebe NM, Del Giudice M, Emery Thompson M, Nickels N, Ponzi D, Zilioli S, Maestripieri D, Gangestad SW. Testosterone, cortisol, and status-striving personality features: a review and empirical evaluation of the Dual Hormone hypothesis. Horm Behav. 2019;109:25–37.

    CAS  PubMed  Google Scholar 

  22. Noguera JC, Aira M, Pérez-Losada M, Domínguez J, Velando A. Glucocorticoids modulate gastrointestinal microbiome in a wild bird 2018.

  23. Antwis RE, Edwards KL, Unwin B, Walker SL, Shultz S. Rare gut microbiota associated with breeding success, hormone metabolites and ovarian cycle phase in the critically endangered eastern black rhino. Microbiome. 2019;7:1–12.

    Google Scholar 

  24. Uren Webster TM, Rodriguez-Barreto D, Consuegra S, Garcia de Leaniz C. Cortisol-related signatures of stress in the fish microbiome. Front Microbiol 2020; 11.

  25. Shi Z, Rothrock MJ, Ricke SC. Applications of microbiome analyses in alternative poultry broiler production systems. Front Vet Sci. 2019;6:157.

    PubMed  PubMed Central  Google Scholar 

  26. Mudd AT, Berding K, Wang M, Donovan SM, Dilger RN. Serum cortisol mediates the relationship between fecal Ruminococcus and brain N-acetylaspartate in the young pig. Gut Microbes. 2017;8:589–600.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ridlon JM, Ikegawa S, Alves JMP, Zhou B, Kobayashi A, Iida T, Mitamura K, Tanabe G, Serrano M, De Guzman A, Cooper P, Buck GA, et al. Clostridium scindens: a human gut microbe with a high potential to convert glucocorticoids into androgens. J Lipid Res. 2013;54:2437–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Moosa Y, Kwon D, de Oliveira T, Wong EB. Determinants of vaginal microbiota composition. Front Cell Infect Microbiol 2020; 10.

  29. Dunlop AL, Mulle JG, Ferranti EP, Edwards S, Dunn AB, Corwin EJ. Maternal microbiome and pregnancy outcomes that impact infant health: a review. Adv Neonatal Care. 2015;15:377–85.

    PubMed  PubMed Central  Google Scholar 

  30. Zhang L, Li C, Zhai Y, Feng L, Bai K, Zhang Z, Huang Y, Li T, Li D, Li H, Cui P, Chen D, et al. Analysis of the vaginal microbiome of giant pandas using metagenomics sequencing. MicrobiologyOpen. 2020;9:1–21.

    Google Scholar 

  31. Javurek AB, Spollen WG, Ali AMM, Johnson SA, Lubahn DB, Bivens NJ, Bromert KH, Ellersieck MR, Givan SA, Rosenfeld CS. Discovery of a novel seminal fluid microbiome and influence of estrogen receptor alpha genetic status. Sci Rep 2016.

  32. Farahani L, Tharakan T, Yap T, Ramsay JW, Jayasena CN, Minhas S. The semen microbiome and its impact on sperm function and male fertility: a systematic review and meta-analysis. Andrology 2020:1–30.

  33. Altmäe S, Franasiak JM, Mändar R. The seminal microbiome in health and disease. Nat Rev Urol. 2019;16:703–21.

    PubMed  Google Scholar 

  34. Weng SL, Chiu CM, Lin FM, Huang WC, Liang C, Yang T, Yang TL, Liu CY, Wu WY, Chang YA, Chang TH, Da Huang H. Bacterial communities in semen from men of infertile couples: Metagenomic sequencing reveals relationships of seminal microbiota to semen quality. PLoS One. 2014;9:110152.

    Google Scholar 

  35. Herkrath CE, Moore IT, Belden LK, Hawley DM, Walters JR, Hopkins WA, Mcglothlin JW. Reproductive physiology, avian malaria, and the cloacal microbiome in tropical Rufous-collared Sparrows (Zonotrichia capensis). 2015.

  36. Escallón C, Belden LK, Moore IT. The cloacal microbiome changes with the breeding season in a wild bird. Integr Org Biol 2019; 1.

  37. Mallott EK, Borries C, Koenig A, Amato KR, Lu A. Reproductive hormones mediate changes in the gut microbiome during pregnancy and lactation in Phayre’s leaf monkeys. Sci Rep. 2020;10:1–9.

    Google Scholar 

  38. Shi Y, Miao ZY, Su JP, Wasser SK. Shift of maternal gut microbiota of tibetan antelope (Pantholops hodgsonii) During the Periparturition Period. Curr Microbiol 2021:1–12.

  39. Miller EA, Beasley DAE, Dunn RR, Archie EA. Lactobacilli dominance and vaginal pH: why is the human vaginal microbiome unique? Front Microbiol. 2016;7:1936.

    PubMed  PubMed Central  Google Scholar 

  40. Shawkey MD, Pillai SR, Hill GE, Siefferman LM, Roberts SR. Bacteria as an agent for change in structural plumage color: Correlational and experimental evidence. Am Nat 2007; 169.

  41. Albone ES, Perry GC. Anal sac secretion of the red fox, Vulpes vulpes; volatile fatty acids and diamines: Implications for a fermentation hypothesis of chemical recognition. J Chem Ecol. 1976;2:101–11.

    CAS  Google Scholar 

  42. Walker-Bolton AD, Parga JA. ‘Stink flirting’ in ring-tailed lemurs (Lemur catta): Male olfactory displays to females as honest, costly signals. Am J Primatol. 2017;79:e22724.

    Google Scholar 

  43. Ezenwa VO, Williams AE. Microbes and animal olfactory communication: where do we go from here? BioEssays. 2014;36:847–54.

    PubMed  Google Scholar 

  44. Carthey AJR, Gillings MR, Blumstein DT. The extended genotype: microbially mediated olfactory communication. Trends Ecol Evol. 2018;33:885–94.

    PubMed  Google Scholar 

  45. Drea CM. Design, delivery and perception of condition-dependent chemical signals in strepsirrhine primates: Implications for human olfactory communication. Philos Trans R Soc B Biol Sci 2020; 375.

  46. Eisenberg JF, Kleiman DG. Olfactory communication in mammals. Annr Rev Ecol System 2003; 3:1–32.

  47. Leclaire S, Jacob S, Greene LK, Dubay GR, Drea CM. Social odours covary with bacterial community in the anal secretions of wild meerkats. Sci Reports. 2017;71(7):1–13.

    Google Scholar 

  48. Theis KR, Venkataraman A, Dycus JA, Koonter KD, Schmitt-Matzen EN, Wagner AP, Holekamp KE, Schmidt TM. Symbiotic bacteria appear to mediate hyena social odors. Proc Natl Acad Sci. 2013;110:19832–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhu J, Arena S, Spinelli S, Liu D, Zhang G, Wei R, Cambillau C, Scaloni A, Wang G, Pelosi P. Reverse chemical ecology: olfactory proteins from the giant panda and their interactions with putative pheromones and bamboo volatiles. Proc Natl Acad Sci U S A. 2017;114:E9802–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Noonan MJ, Tinnesand HV, Müller CT, Rosell F, Macdonald DW, Buesching CD. Knowing me, knowing you: anal gland secretion of European Badgers (Meles meles) codes for individuality, sex and social group membership. J Chem Ecol. 2019;45:823–37.

    CAS  PubMed  Google Scholar 

  51. Greene LK, Bornbusch SL, McKenney EA, Harris RL, Gorvetzian SR, Yoder AD, Drea CM. The importance of scale in comparative microbiome research: New insights from the gut and glands of captive and wild lemurs. Am J Primatol 2019; 81:e22974.

  52. Li D, Chen B, Zhang L, Gaur U, Ma T, Jie H, Zhao G, Wu N, Xu Z, Xu H, Yao Y, Lian T, et al. The musk chemical composition and microbiota of Chinese forest musk deer males. Sci Rep. 2016;6:1–10.

    Google Scholar 

  53. Maraci Ö, Engel K, Caspers BA. Olfactory communication via microbiota: what is known in birds? Genes (Basel) 2018; 9.

  54. Grieves LA, Gloor GB, Bernards MA, MacDougall-Shackleton EA. Songbirds show odour-based discrimination of similarity and diversity at the major histocompatibility complex. Anim Behav. 2019;158:131–8.

    Google Scholar 

  55. Brunetti AE, Lyra ML, Melo WGP, Andrade LE, Palacios-Rodríguez P, Prado BM, Haddad CFB, Pupo MT, Lopes NP. Symbiotic skin bacteria as a source for sex-specific scents in frogs. Proc Natl Acad Sci. 2019;116:2124–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Smith CC, Mueller UG. Sexual transmission of beneficial microbes. Trends Ecol Evol. 2015;30:438–40.

    PubMed  Google Scholar 

  57. Uchihashi M, Bergin IL, Bassis CM, Hashway SA, Chai D, Bell JD. Influence of age, reproductive cycling status, and menstruation on the vaginal microbiome in baboons (Papio anubis ). Am J Primatol. 2015;77:563–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Power ML, Quaglieri C, Schulkin J. Reproductive microbiomes: a new thread in the microbial network. Reprod Sci. 2017;24:1482–92.

    PubMed  Google Scholar 

  59. Heil BA, Paccamonti DL, Sones JL. Role for the mammalian female reproductive tract microbiome in pregnancy outcomes. Physiol Genomics. 2019;51:390–9.

    CAS  PubMed  Google Scholar 

  60. Yildirim S, Yeoman CJ, Janga SC, Thomas SM, Ho M, Leigh SR, Consortium PM, White BA, Wilson BA, Stumpf RM. Primate vaginal microbiomes exhibit species specificity without universal Lactobacillus dominance. ISME J. 2014;8:2431–44.

    Google Scholar 

  61. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SSK, McCulle SL, Karlebach S, Gorle R, Russell J, Tacket CO, Brotman RM, Davis CC, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci. 2011;108:4680–7.

    CAS  PubMed  Google Scholar 

  62. Silva HVR, Silva AR, da Silvada LDM, Comizzoli P. Semen Cryopreservation and Banking for the Conservation of Neotropical Carnivores. Biopreserv Biobank. 2019;17(2):183–8. https://doi.org/10.1089/bio.2018.0104.

    Article  PubMed  Google Scholar 

  63. Zainuddin ZZ, Mohamed Tarmizi MR, Yap KC, Comizzoli P, Sipangkui S. First evaluations and cryopreservation of semen samples from sunda clouded leopards (Neofelis diardi). Animals. 10(6):1072. https://doi.org/10.3390/ani10061072.

    Article  PubMed Central  Google Scholar 

  64. Zainuddin ZZ, Sipangkui S, Farqhan Kelana M, Chee YK, Tarmizi MRM, Comizzoli P. Repeated evaluations of testes and semen characteristics in two binturongs (Arctictis binturong). Front Vet Sci. 2021;8:658573.

    PubMed  PubMed Central  Google Scholar 

  65. Comizzoli P. Biobanking efforts and new advances in male fertility preservation for rare and endangered species. Asian J Androl. 2015;17:640–5.

    PubMed  PubMed Central  Google Scholar 

  66. Baud D, Vulliemoz N, Morales Zapata MV, Greub G, Vouga M, Stojanov M. Waddlia chondrophila and male infertility. Microorganisms. 2020;8:136.

    CAS  PubMed Central  Google Scholar 

  67. Santos CS, Silva AM, Maia KM, Rodrigues GSO, Feijó FMC, Alves ND, Oliveira MF, Silva AR. Composition of semen and foreskin mucosa aerobic microbiota and its impact on sperm parameters of captive collared peccaries ( Pecari tajacu). J Appl Microbiol. 2020;129:521–31.

    CAS  PubMed  Google Scholar 

  68. Fullston T, McPherson NO, Owens JA, Kang WX, Sandeman LY, Lane M. Paternal obesity induces metabolic and sperm disturbances in male offspring that are exacerbated by their exposure to an ‘obesogenic’ diet. Physiol Rep 2015; 3.

  69. Miller EA, Livermore JA, Alberts SC, Tung J, Archie EA. Ovarian cycling and reproductive state shape the vaginal microbiota in wild baboons. Microbiome 2017.

  70. Bornbusch SL, Greene LK, McKenney EA, Volkoff SJ, Midani FS, Joseph G, Gerhard WA, Iloghalu U, Granek J, Gunsch CK. A comparative study of gut microbiomes in captive nocturnal strepsirrhines. Am J Primatol 2019; 81.

  71. Nunn KL, Wang YY, Harit D, Humphrys MS, Ma B, Cone R, Ravel J, Lai SK. Enhanced trapping of hiv-1 by human cervicovaginal mucus is associated with lactobacillus crispatus-dominant microbiota. MBio. 2015;6:1–9.

    Google Scholar 

  72. da Silva GA, Monteiro FOB, Dias HLT, Cavalcante RO, Sampaio AIF, da Conceição MEBAM, Takeshita RSC, de Castro PHG, Feijó FMC, Rahal SC. Qualitative analysis of preputial and vaginal bacterial microbiota in owl monkeys (Aotus azarai infulatus) raised in captivity. J Med Primatol 2013; 42:71–78.

  73. Mändar R, Punab M, Borovkova N, Lapp E, Kiiker R, Korrovits P, Metspalu A, Krjutškov K, Nõlvak H, Preem J-K, Oopkaup K, Salumets A, et al. Complementary seminovaginal microbiome in couples. Res Microbiol. 2015;166:440–7.

    PubMed  Google Scholar 

  74. MacManes MD. Promiscuity in mice is associated with increased vaginal bacterial diversity. Naturwissenschaften. 2011;98:951–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Kenyon CR, Delva W, Brotman RM. Differential sexual network connectivity offers a parsimonious explanation for population-level variations in the prevalence of bacterial vaginosis: A data-driven, model-supported hypothesis. BMC Womens Health 2019; 19.

  76. White J, Richard M, Massot M, Meylan S. Cloacal bacterial diversity increases with multiple mates: evidence of sexual transmission in female common lizards. PLoS ONE. 2011;6:22339.

    Google Scholar 

  77. Schwebke JR, Desmond R. Risk factors for bacterial vaginosis in women at high risk for sexually transmitted diseases. Sex Transm Dis. 2005;32:654–8.

    PubMed  Google Scholar 

  78. Moumne O, Hampe ME, Montoya-Williams D, Carson TL, Neu J, Francois M, Rhoton-Vlasak A, Lemas DJ. Implications of the vaginal microbiome and potential restorative strategies on maternal health: a narrative review. J Perinat Med. 2021;49:402–11.

    PubMed  Google Scholar 

  79. Mesa MD, Loureiro B, Iglesia I, Gonzalez SF, Olivé EL, Algar OG, Solana MJ, Perez MJC, Sainz T, Martinez L, Escuder-Vieco D, Parra-Llorca A, et al. The evolving microbiome from pregnancy to early infancy: a comprehensive review. Nutrients 2020; 12.

  80. Chen C, Song X, Wei W, Zhong H, Dai J, Lan Z, Li F, Yu X, Feng Q, Wang Z, Xie H, Chen X, et al. The microbiota continuum along the female reproductive tract and its relation to uterine-related diseases. Nat Commun 2017.

  81. Theis KR, Romero R, Winters AD, Greenberg JM, Gomez-Lopez N, Alhousseini A, Bieda J, Maymon E, Pacora P, Fettweis JM, Buck GA, Jefferson KK, et al. Does the human placenta delivered at term have a microbiota? Results of cultivation, quantitative real-time PCR, 16S rRNA gene sequencing, and metagenomics. Am J Obstet Gynecol. 2019;220:267.e1-267.e39.

    CAS  Google Scholar 

  82. Gil A, Rueda R, Ozanne SE, van der Beek EM, van Loo-Bouwman C, Schoemaker M, Marinello V, Venema K, Stanton C, Schelkle B, Flourakis M, Edwards CA. Is there evidence for bacterial transfer via the placenta and any role in the colonization of the infant gut?–a systematic review. Crit Rev Microbiol. 2020;46:493–507.

    CAS  PubMed  Google Scholar 

  83. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med 2014; 6.

  84. Parnell LA, Briggs CM, Cao B, Delannoy-Bruno O, Schrieffer AE, Mysorekar IU. Microbial communities in placentas from term normal pregnancy exhibit spatially variable profiles. Sci Rep. 2017;7:1–11.

    Google Scholar 

  85. Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep. 2016;6:1–13.

    Google Scholar 

  86. Lim ES, Rodriguez C, Holtz LR. Amniotic fluid from healthy term pregnancies does not harbor a detectable microbial community. Microbiome 2018; 6.

  87. Rehbinder EM, Lødrup Carlsen KC, Staff AC, Angell IL, 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.e1-289.e12.

    Google Scholar 

  88. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. A critical assessment of the ‘sterile womb’ and ‘in utero colonization’ hypotheses: Implications for research on the pioneer infant microbiome. Microbiome. 2017;5:1–19.

    Google Scholar 

  89. Mols KL, Boe-Hansen GB, Mikkelsen D, Bryden WL, Cawdell-Smith AJ. Prenatal establishment of the foal gut microbiota: a critique of the in utero colonisation hypothesis. Anim Prod Sci. 2020;60:2080–92.

    Google Scholar 

  90. Vidal S, Kegler K, Posthaus H, Perreten V, Rodriguez-Campos S. Amplicon sequencing of bacterial microbiota in abortion material from cattle. Vet Res. 2017;48:64.

    PubMed  PubMed Central  Google Scholar 

  91. Husso A, Lietaer L, Pessa-Morikawa T, Grönthal T, Govaere J, Van Soom A, Iivanainen A, Opsomer G, Niku M. The composition of the microbiota in the full-term fetal gut and amniotic fluid: a bovine cesarean section study. Front Microbiol 2021; 12.

  92. Parris KM, Amabebe E, Cohen MC, Anumba D. Placental microbial-metabolite profiles and inflammatory mechanisms associated with preterm birth. J Clin Pathol. 2021;74:10–8.

    CAS  PubMed  Google Scholar 

  93. Kimura I, Miyamoto J, Ohue-Kitano R, Watanabe K, Yamada T, Onuki M, Aoki R, Isobe Y, Kashihara D, Inoue D, Inaba A, Takamura Y, et al. Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science (80- ) 2020; 367.

  94. Sarkar A, Yoo JY, Dutra SVO, Morgan KH, Groer M. The association between early-life gut microbiota and long-term health and diseases. J Clin Med. 2021;10:459.

    PubMed  PubMed Central  Google Scholar 

  95. Campbell JM, Lane M, Owens JA, Bakos HW. Paternal obesity negatively affects male fertility and assisted reproduction outcomes: a systematic review and meta-analysis. Reprod Biomed Online. 2015;31:593–604.

    PubMed  Google Scholar 

  96. Kumbhare S V, Patangia D V, Patil RH, Shouche YS, Patil NP. Review Factors influencing the gut microbiome in children: from infancy to childhood 2038.

  97. Stinson LF, Payne MS, Keelan JA. A critical review of the bacterial baptism hypothesis and the impact of cesarean delivery on the infant microbiome. Front Med. 2018;5:1.

    Google Scholar 

  98. Shaterian N, Abdi F, Ghavidel N, Alidost F. Role of cesarean section in the development of neonatal gut microbiota: a systematic review. Open Med 2021; 16.

  99. Trevelline BK, Fontaine SS, Hartup BK, Kohl KD. Conservation biology needs a microbial renaissance: A call for the consideration of host-associated microbiota in wildlife management practices. Proc R Soc B Biol Sci 2019; 286.

  100. Weiss S, Taggart D, Smith I, Helgen K, Eisenhofer R. Host Reproductive Cycle Influences the Pouch Microbiota of Wild Southern Hairy-Nosed Wombats (Lasiorhinus Latifrons) 2020:1–18.

  101. Muletz-Wolz CR, Kurata NP, Himschoot EA, Wenker ES, Quinn EA, Hinde K, Power ML, Fleischer RC. Diversity and temporal dynamics of primate milk microbiomes. Am J Primatol. 2019;81:e22994.

    PubMed  PubMed Central  Google Scholar 

  102. LaTuga MS, Stuebe A, Seed PC. A review of the source and function of microbiota in breast milk. Semin Reprod Med. 2014;32:68–73.

    PubMed  Google Scholar 

  103. Heikkilä MP, Saris PEJ. Inhibition of Staphylococcus aureus by the commensal bacteria of human milk. J Appl Microbiol. 2003;95:471–8.

    PubMed  Google Scholar 

  104. Nunez N, Réot L, Menu E. Neonatal immune system ontogeny: the role of maternal microbiota and associated factors. How might the non-human primate model enlighten the path? Vaccines. 2021;9:584.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Gillespie MJ, Stanley D, Chen H, Donald JA, Nicholas KR, Moore RJ, Crowley TM. Functional Similarities between Pigeon ‘Milk’ and Mammalian Milk: Induction of Immune Gene Expression and Modification of the Microbiota. PLoS One 2012; 7.

  106. Jacquin L, Blottière L, Haussy C, Perret S, Gasparini J. Prenatal and postnatal parental effects on immunity and growth in ‘lactating’ pigeons. Funct Ecol. 2012;26:866–75.

    Google Scholar 

  107. Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science (80-). 2016;352:539–44.

    CAS  Google Scholar 

  108. McKenzie VJ, Song SJ, Delsuc F, Prest TL, Oliverio AM, Korpita TM, Alexiev A, Amato KR, Metcalf JL, Kowalewski M, Avenant NL, Link A, et al. The effects of captivity on the mammalian gut microbiome. Integrative and Comparative Biology, vol. 57. Oxford University Press; 2017:690–704.

  109. Dominguez Bello MG, Knight R, Gilbert JA, Blaser MJ. Preserving microbial diversity. Microbiota. 2018;362:6410.

    Google Scholar 

  110. Banerjee A, Cornejo J, Bandopadhyay R. Emergent climate change impact throughout the world: call for ‘Microbiome Conservation’ before it’s too late. Biodivers Conserv. 2020;29:345–8.

    Google Scholar 

  111. Sun CH, Liu HY, Liu B, Yuan BD, Lu CH. Analysis of the gut microbiome of wild and captive Père David’s deer. Front Microbiol 2019; 10.

  112. Rivera AJ, Frank JA, Stumpf R, Salyers AA, Wilson BA, Olsen GJ, Leigh S. Differences between the normal vaginal bacterial community of baboons and that of humans. Am J Primatol. 2011;73:119–26.

    PubMed  Google Scholar 

  113. Jašarević E, Howerton CL, Howard CD, Bale TL. Alterations in the vaginal microbiome by maternal stress are associated with metabolic reprogramming of the offspring gut and brain. Endocrinology. 2015;156:3265–76.

    PubMed  PubMed Central  Google Scholar 

  114. Amabebe E, Anumba DOC. Female gut and genital tract microbiota-induced crosstalk and differential effects of short-chain fatty acids on immune sequelae. Front Immunol. 2020;11:2184.

    CAS  PubMed  PubMed Central  Google Scholar 

  115. van Leeuwen P, Mykytczuk N, Mastromonaco GF, Schulte-Hostedde AI. Effects of captivity, diet, and relocation on the gut bacterial communities of white-footed mice. Ecol Evol. 2020;10:4677–90.

    PubMed  PubMed Central  Google Scholar 

  116. Bornbusch SL, Harris RL, Grebe NM, Roche K, Dimac-Stohl K, Drea CM. Longitudinal effects of antibiotics and fecal transplant on lemur gut microbiota structure, associations, and resistomes. bioRxiv 2020:2020.11.11.378349.

  117. Lemas DJ, Yee S, Cacho N, Miller D, Cardel M, Gurka M, Janicke D, Shenkman E. Exploring the contribution of maternal antibiotics and breastfeeding to development of the infant microbiome and pediatric obesity. Semin Fetal Neonatal Med. 2016;21:406–9.

    PubMed  Google Scholar 

  118. Yang L, Hao Y, Hu J, Kelly D, Li H, Brown S, Tasker C, Roche NE, Chang TL, Pei Z. Differential effects of depot medroxyprogesterone acetate administration on vaginal microbiome in Hispanic White and Black women. Emerg Microb Infect. 2019;8:197–210.

    CAS  Google Scholar 

  119. Brooks JP, Edwards DJ, Blithe DL, Fettweis JM, Serrano MG, Sheth NU, Strauss JF, Buck GA, Jefferson KK. Effects of combined oral contraceptives, depot medroxyprogesterone acetate and the levonorgestrel-releasing intrauterine system on the vaginal microbiome. Contraception. 2017;95:405–13.

    CAS  PubMed  Google Scholar 

  120. Crawford JC, Boulet M, Drea CM. Smelling wrong: hormonal contraception in lemurs alters critical female odour cues. Proc R Soc B Biol Sci. 2011;278:122–30.

    CAS  Google Scholar 

  121. Roberts SC, Gosling LM, Carter V, Petrie M. MHC-correlated odour preferences in humans and the use of oral contraceptives. Proc R Soc B Biol Sci. 2008;275:2715–22.

    Google Scholar 

  122. Kuukasjärvi S, Eriksson CJP, Koskela E, Mappes T, Nissinen K, Rantala MJ. Attractiveness of women’s body odors over the menstrual cycle: the role of oral contraceptives and receiver sex. Behav Ecol. 2004;15:579–84.

    Google Scholar 

  123. Hayes TB, Khoury V, Narayan A, Nazir M, Parka A, Brown T, Adame L, Chan E, Buchholz D, Stueve T, Gallipeau S. Atrazine induces complete feminization and chemical castration in male African clawed frogs (Xenopus laevis). Proc Natl Acad Sci U S A. 2010;107:4612–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Zhang W, Guo R, Ai S, Yang Y, Ding J, Zhang Y. Long-term heavy metal pollution varied female reproduction investment in free-living anura Bufo raddei. Ecotoxicol Environ Saf. 2018;159:136–42.

    CAS  PubMed  Google Scholar 

  125. Nielsen LN, Roager HM, Casas ME, Frandsen HL, Gosewinkel U, Bester K, Licht TR, Hendriksen NB, Bahl MI. Glyphosate has limited short-term effects on commensal bacterial community composition in the gut environment due to sufficient aromatic amino acid levels. Environ Pollut. 2018;233:364–76.

    CAS  PubMed  Google Scholar 

  126. Mesnage R, Teixeira M, Mandrioli D, Falcioni L, Ducarmon QR, Zwittink RD, Mazzacuva F, Caldwell A, Halket J, Amiel C, Panoff JM, Belpoggi F, et al. Use of shotgun metagenomics and metabolomics to evaluate the impact of glyphosate or roundup mon 52276 on the gut microbiota and serum metabolome of sprague-dawley rats. Environ Health Perspect. 2021;129:1–15.

    Google Scholar 

  127. Jiménez RR, Alvarado G, Ruepert C, Ballestero E, Sommer S. The fungicide chlorothalonil changes the amphibian skin microbiome: a potential factor disrupting a host disease-protective trait. Appl Microbiol. 2021;1:26–37.

    Google Scholar 

  128. Chiu K, Warner G, Nowak RA, Flaws JA, Mei W. The impact of environmental chemicals on the gut microbiome. Toxicol Sci. 2020;176:253–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Claus SP, Guillou H, Ellero-Simatos S. The gut microbiota: a major player in the toxicity of environmental pollutants? Npj Biofilms Microbiomes. 2016;2:1–11.

    Google Scholar 

  130. Ruuskanen S, Rainio MJ, Gómez-Gallego C, Selenius O, Salminen S, Collado MC, Saikkonen K, Saloniemi I, Helander M. Glyphosate-based herbicides influence antioxidants, reproductive hormones and gut microbiome but not reproduction: A long-term experiment in an avian model. Environ Pollut 2020; 266.

  131. Syromyatnikov MY, Isuwa MM, Savinkova OV, Derevshchikova MI, Popov VN. The effect of pesticides on the microbiome of animals. Agric. 2020;10:79.

    CAS  Google Scholar 

  132. Baquero F, Martínez JL, Cantón R. Antibiotics and antibiotic resistance in water environments. Curr Opin Biotechnol. 2008;19:260–5.

    CAS  PubMed  Google Scholar 

  133. Ramey AM, Ahlstrom CA. Antibiotic resistant bacteria in wildlife: Perspectives on trends, acquisition and dissemination, data gaps, and future directions. J Wildl Dis. 2020;56:1–15.

    PubMed  Google Scholar 

  134. Martinez JL. Environmental pollution by antibiotics and by antibiotic resistance determinants. Environ Pollut. 2009;157:2893–902.

    CAS  PubMed  Google Scholar 

  135. Sanderson H, Fricker C, Brown RS, Majury A, Liss SN. Antibiotic resistance genes as an emerging environmental contaminant. Environ Rev. 2016;24:205–18.

    Google Scholar 

  136. SL B, RL H, NM G, K R, K D-S, CM D. Longitudinal effects of antibiotics and fecal transplant on lemur gut microbiota structure, associations, and resistomes 2020.

  137. Robinson TP, Bu DP, Carrique-Mas J, Fèvre EM, Gilbert M, Grace D, Hay SI, Jiwakanon J, Kakkar M, Kariuki S, Laxminarayan R, Lubroth J, et al. Antibiotic resistance is the quintessential One Health issue. Trans R Soc Trop Med Hyg. 2016;110:377–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Bornbusch SL, Drea CM. Antibiotic resistance genes in lemur gut and soil microbiota along a gradient of anthropogenic disturbance. Front Ecol Evol 1AD; 0:514.

  139. Duffield KR, Hampton KJ, Houslay TM, Hunt J, Rapkin J, Sakaluk SK, Sadd BM. Age-dependent variation in the terminal investment threshold in male crickets. Evolution (N Y). 2018;72:578–89.

    Google Scholar 

  140. Williams GC. Natural selection, the costs of reproduction, and a refinement of lack’s principle. Am Natural. 2015;100:687–90.

    Google Scholar 

  141. Brannelly LA, Webb R, Skerratt LF, Berger L. Amphibians with infectious disease increase their reproductive effort: Evidence for the terminal investment hypothesis. Open Biol 2016; 6.

  142. Kelleher SR, Scheele BC, Silla AJ, Keogh JS, Hunter DA, Endler JA, Byrne PG. Disease influences male advertisement and mating outcomes in a critically endangered amphibian. Anim Behav. 2021;173:145–57.

    Google Scholar 

  143. Pigeault R, Cozzarolo CS, Choquet R, Strehler M, Jenkins T, Delhaye J, Bovet L, Wassef J, Glaizot O, Christe P. Haemosporidian infection and co-infection affect host survival and reproduction in wild populations of great tits. Int J Parasitol. 2018;48:1079–87.

    CAS  PubMed  Google Scholar 

  144. Velando A, Drummond H, Torres R. Senescent birds redouble reproductive effort when ill: confirmation of the terminal investment hypothesis. Proc R Soc B Biol Sci. 2006;273:1443–8.

    Google Scholar 

  145. Bonneaud C, Mazuc J, Chastel O, Westerdahl H, Sorci G. Terminal investment induced by immune challenge and fitness traits associated with major histocompatibility complex in the house sparrow. Evolution (N Y). 2004;58:2823–30.

    CAS  Google Scholar 

  146. Names GR, Krause JS, Schultz EM, Angelier F, Parenteau C, Ribout C, Hahn TP, Wingfield JC. Relationships between avian malaria resilience and corticosterone, testosterone and prolactin in a Hawaiian songbird. Gen Comp Endocrinol. 2021;308:113784.

    CAS  PubMed  Google Scholar 

  147. Davis NM, Proctor DM, Holmes SP, Relman DA, Callahan BJ. Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome. 2018;6:1–14.

    Google Scholar 

  148. Comizzoli P, Ann OM. Understanding reproductive aging in wildlife to improve animal conservation and human reproductive health. Front Cell Dev Biol. 2021;9:1281.

    Google Scholar 

  149. Holtze S, Gorshkova E, Braude S, Cellerino A, Dammann P, Hildebrandt TB, Hoeflich A, Hoffmann S, Koch P, Tozzini ET, Skulachev M, Skulachev VP, et al. Alternative animal models of aging research. Front Mol Biosci. 2021;8:660959.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Comizzoli P. New directions to understand and learn from embryonic diapause in mammals. 2020. https://doi.org/10.1530/biosciprocs.10.016.

  151. Jin Song S, Woodhams DC, Martino C, Allaband C, Mu A, Javorschi-Miller-Montgomery S, Suchodolski JS, Knight R. Engineering the microbiome for animal health and conservation. Exp Biol Med. 2019;244:494–504.

    CAS  Google Scholar 

  152. Ning D, Yuan M, Wu L, Zhang Y, Guo X, Zhou X, Yang Y, Arkin AP, Firestone MK, Zhou J. A quantitative framework reveals ecological drivers of grassland microbial community assembly in response to warming. Nat Commun. 2020;11:1–12.

    Google Scholar 

  153. Miller ET, Svanbäck R, Bohannan BJM. Microbiomes as metacommunities: understanding host-associated microbes through metacommunity ecology. Trends Ecol Evol. 2018;33:926–35.

    PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

PC, PM, SL, and CRMW conceptualized the manuscript, wrote the first draft, and edited subsequent versions. All authors contributed ideas and commented on the text. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Pierre Comizzoli.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Comizzoli, P., Power, M.L., Bornbusch, S.L. et al. Interactions between reproductive biology and microbiomes in wild animal species. anim microbiome 3, 87 (2021). https://doi.org/10.1186/s42523-021-00156-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s42523-021-00156-7

Keywords