Next Article in Journal
The Global Epidemiology of Bovine Leukemia Virus: Current Trends and Future Implications
Next Article in Special Issue
Programmed Grooming after 30 Years of Study: A Review of Evidence and Future Prospects
Previous Article in Journal
Pathogenic Potential of Coagulase-Positive Staphylococcus Strains Isolated from Aviary Capercaillies and Free-Living Birds in Southeastern Poland
Previous Article in Special Issue
Evaluating the Presence of Disgust in Animals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neurobiology of Pathogen Avoidance and Mate Choice: Current and Future Directions

by
Dante Cantini
1,
Elena Choleris
1 and
Martin Kavaliers
1,2,*
1
Department of Psychology, College of Social and Applied Human Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
2
Department of Psychology, Western University, London, ON N6A 3K7, Canada
*
Author to whom correspondence should be addressed.
Animals 2024, 14(2), 296; https://doi.org/10.3390/ani14020296
Submission received: 27 November 2023 / Revised: 9 January 2024 / Accepted: 13 January 2024 / Published: 17 January 2024
(This article belongs to the Special Issue Interrelationships between Animal Behaviors and Control of Diseases)

Abstract

:

Simple Summary

The risk of parasitic infection has a major influence on animal behaviour. Organisms must adjust their behaviour to avoid various modes of parasitic infection and pathogen acquisition. Social species are at an increased risk of parasitic transmission as they spend more time in the proximity of others that may carry parasites. The detection of parasitic risk is also critical in mate assessment and choice. Perceptual systems and behavioural responses have evolved to detect individuals who are parasitized and pose the risk of parasitic transmission. This includes the integration of inputs from various sensory modalities (e.g., olfaction), brain regions and networks, and neuromodulatory systems. Understanding the neurobiological systems involved in detecting the parasite infection risk and the expression of disgust will allow us to better understand the evolution and regulation of pathogen avoidance and mate choice.

Abstract

Animals are under constant threat of parasitic infection. This has influenced the evolution of social behaviour and has strong implications for sexual selection and mate choice. Animals assess the infection status of conspecifics based on various sensory cues, with odours/chemical signals and the olfactory system playing a particularly important role. The detection of chemical cues and subsequent processing of the infection threat that they pose facilitates the expression of disgust, fear, anxiety, and adaptive avoidance behaviours. In this selective review, drawing primarily from rodent studies, the neurobiological mechanisms underlying the detection and assessment of infection status and their relations to mate choice are briefly considered. Firstly, we offer a brief overview of the aspects of mate choice that are relevant to pathogen avoidance. Then, we specifically focus on the olfactory detection of and responses to conspecific cues of parasitic infection, followed by a brief overview of the neurobiological systems underlying the elicitation of disgust and the expression of avoidance of the pathogen threat. Throughout, we focus on current findings and provide suggestions for future directions and research.

1. Introduction

For many species, living in social groups provides evolutionary benefits, such as protection from predators, sharing of information and resources, and increased potential mating opportunities [1,2,3,4,5]. Living in large social groups also has costs and puts individuals at an increased risk of infection [6,7,8]. There is evidence that a larger social group size positively correlates with an increased individual risk of disease and parasitic infection, largely attributed to an increased contact rate among individuals [8,9,10,11] (for further discussions, see Altizer et al. (2003) [9]). Contrary to the view of parasitic risk being a cost of social groups, it is speculated that group living may confer anti-parasitic benefits via resistance to parasites (i.e., social immunity) and enhanced tolerance of parasitic infection (i.e., social interactions that improve physical condition via increased resource acquisition or social support) [10,12]. Nonetheless, the risk of parasitic infection via conspecific transmission is an integral aspect of group living that shapes the evolution of sociality.
In the wild, individuals of most social and non-social species harbour varying levels of parasites and pathogens, including harmful bacteria and viruses. For example, members of natural mouse populations frequently carry some form of parasite, such as skin mites and various nematode species [13,14]. Gregory et al. (1992) [14] proposed that approximately 80% of all wild wood mice are likely to be infected with a parasitic load of 10 to 25 helminth parasites. In natural environments, as opposed to most sterile laboratory environments, it is unlikely to find organisms that are fully parasite-free [15]. Here, we will refer to parasites broadly to encapsulate any relationship in which one organism (the parasite) benefits by living in or on the host organism at the cost of the host [16].
The prevalence of parasites in wild populations puts evolutionary pressure on individuals to assess and avoid infection risk. Detection and avoidance of parasites is beneficial for individuals at both the proximate level (avoiding direct transmission of parasites and disease to oneself) and the ultimate level (selection of genes that will improve the disease resistance of one’s offspring). To avoid infection, animals must modulate their behaviour and interactions with conspecifics based on social information that conveys the health status of others.
Curtis and Biran (2001) [17] proposed disgust as an evolutionary adaptation that biases behaviour toward the avoidance of parasitic infection, contaminants, and toxin risk. Disgust is proposed as a fundamental affective/emotional state that underlies and facilitates the behavioural avoidance of individuals that are perceived on the basis of various sensory cues that represent a potential risk of pathogen exposure and infection [18,19,20]. In addition, information regarding infection or contagion can elicit other forms of affect, such as fear and anxiety [19,21].
It has been further proposed that individuals face a ‘landscape of disgust’, in which animals must detect ‘mountains’ of high infection risk and ‘valleys’ of low infection risk [22]. The ecological and evolutionary consequences of an individual’s responses have been framed in terms of the ecology of disgust [19]. This is particularly pertinent for social behaviours, including mate choice and partner assessment. Individuals who are better at processing sensory information, which leads to appropriate disgust responses and elicitation of avoidance, are less likely to come into contact with parasites, which reduces their probability of infection.
In this selective review, we will consider aspects of the neurobiological behavioural mechanisms underlying parasite detection and avoidance and their implications for mate choice in rodents. We will initially provide a brief overview of aspects of (i) mate choice relevant to pathogen avoidance. Then, we will specifically focus on (ii) the olfactory detection of and response to conspecific cues of parasitic infection and immune status. Finally, (iii) the neurobiology underlying the elicitation of disgust and expression of avoidance and aversive responses to pathogen threat (see Figure 1 for a visual representation of the topics covered in this review). Focusing on select current findings, we will provide suggestions for future research.

2. Aspects of Mate Choice Relevant to Pathogen Avoidance

2.1. Mate Choice

Mice and many other rodents have a polygamous mating system, meaning that both males and females will mate with multiple different partners throughout their reproductive lifespan [23]. A polygamous mating system makes these rodents particularly susceptible to infection by contagious parasites due to the increased frequency of physical proximity and sexual contact with multiple individuals [9,19]. Appropriate assessment and detection of parasitic infection in conspecifics is especially pertinent during mate choice, as there can be a direct risk to the individual through sexually transmitted parasites, such as the protozoan parasite, Toxoplasma gondii [24]. The risk of infection creates a strong selective pressure on the ability to detect actually and potentially parasitized individuals. Many vertebrates, including rodents, use odour cues to detect parasitic infection, and these odours also function as olfactory phenotypes that inform mate choice e.g., [25,26,27,28,29]. Phenotypic secondary sexual characteristics that are generally used to assess a potential mate’s viability can also serve as an indication of the individual’s parasitic load; this has been termed the ‘contagion indicator’ hypothesis [30].
The strong selective pressure to appropriately assess a potential mate’s infection status has both direct and indirect evolutionary benefits for the individual [24,30,31]. Loehle (1995) [32] theorized that females are more risk-averse to disease threats. As a result, female aversion to parasitized males has likely evolved to both avoid direct transmission of contagious pathogens during mating and select for heritable parasite resistance [24,30]. Thus, the selection of non-parasitized mates is beneficial at both the proximate level (avoiding direct transmission of parasites to oneself) and the ultimate level (selection of genes that will improve the disease resistance of one’s offspring). Evidence to support the ultimate-level benefits is provided by the finding that female mice that chose males with ‘maximal’ immune function produced offspring with more robust immune systems [33]. As such, the ability to recognize and avoid infected, or potentially infected, individuals is a key component of mate choice.
In rodents, and most other mammals, sexual behaviour can be divided into two phases: the appetitive (precopulatory) and consummatory (copulatory) phases [34]. The appetitive phase involves the motivation to approach and engage in an assessment of a potential mate [35]. This has also been considered in terms of incentive salience and the rewarding value of the potential mate [36,37,38]. Mate choice, which encompasses the appetitive phase, can be divided into two categories: preference (the preferential order of attractiveness in which an individual ranks potential mates) and choosiness (the responsiveness to potential mates and the amount of effort expended to assess the mate and make the choice) [19,39]. In theory, if all potential mates are available, the chosen mate should closely reflect the peak mate preference. Within the context of parasitic infection, if an uninfected versus an infected mate is available to a healthy individual, the uninfected mate is generally preferred [40]. However, preference and mate choice are not always mutually exclusive. It should be noted that some researchers believe that choice can only be said to have occurred after mating. Preferences are inferred from choices that are made using a proxy (e.g., odours) for mate preferences. For a detailed consideration of the design of mate choice experiments and the use/measure of choices and preferences, see Dougherty (2020) [41] and Dougherty (2023) [42] and the tables and references therein.
Patterns of mate choice can be altered by changing the costs of choosiness without altering the mate preference (i.e., without changing who would be the most preferable mate). As indicated, in the wild, as opposed to laboratory conditions, there may not be a direct choice but rather potential sequential encounters or odour exposures. This constraint on choosiness results in uncertainty regarding the condition and infection status of subsequent potential mates and their reward value. Indeed, the results of studies with rodents in semi-natural environments have suggested that female copulation and impregnation do not necessarily correspond to perceived female mate preference [43]. In a semi-naturalistic study on rats, Chu and Ågmo (2014) [43] demonstrated that when the female preference was defined by the frequency of female sniffing of the male (a behaviour typically associated with preference in most classical laboratory studies), there was no difference in sexual behaviour among the preferred and non-preferred males, and thus female preference within these parameters did not impact male reproductive success. This suggests that in naturalistic conditions, a preferred male may not produce more offspring than a non-preferred male.
Therefore, in nature, not all individuals have the choice to mate with the preferred individual, and thus mate choice is not always representative of mate preference. As a result, female preference for non-parasitized individuals is not always evident and is influenced by other variables. Female mice that are pre-exposed to the odour of parasitized males do not show an initial preference for non-parasitized male odour [26]. Additionally, it was found that a parasitized male odour paired with the odour of an estrous female attenuated the aversive response and resulted in increased female preference for the parasitized male odour [44]. Individuals pay attention to the social and mate decisions of conspecifics and use them to determine their own choices, displaying mate choice copying [45,46]. These findings suggest that the preference for healthy mates can be influenced by the social context and indirect social information that results in ‘mate choice copying’, negating the notion that initial preference will always be directed to the non-parasitized potential mate [46,47]. Whether or not mate choice copying is utilized to avoid potentially infected individuals remains to be determined. However, there is evidence that female mice will avoid the odours of uninfected males that have been associated with those of infected males [48].
The influence of the parasitic infection risk on sexual behaviour and mate choice heavily influences sexual selection [40,49]. Despite some notable exceptions, animals generally demonstrate preference and choice for parasite-free mates to increase both direct and indirect fitness [50,51,52,53,54]. To adaptively reproduce, animals must engage in appropriate behavioural responses when faced with potential infection risks. These behavioural responses not only influence the transfer of genes in a population but also serve to protect the individual against parasitism as well as mitigate the pervasiveness of parasites at the population level. As we briefly discuss in the following sections, the affective disgust response is integral to the behavioural responses that influence mate choice regarding the parasitic infection risk.

2.2. Approach and Avoidance

Loehle (1995) [32] postulated that organisms may employ ‘social barriers’ to mitigate the infection risk, suggesting that various social behaviours (e.g., general social avoidance and avoidance of mating with infected individuals) may effectively reduce pathogen transmission. Hamilton and Zuk (1982) [55] were among the first to directly hypothesize that animals benefit from inspecting a conspecific’s urinary and fecal odours to select a parasite-free or resistant mate. The assessment of infection status facilitates the expression of the appropriate adaptive social response directed toward the conspecific. The process underlying the social assessment of conspecifics involves social cognition, which includes the acquisition of social information that is used to assess the status of other individuals [19,56]. Social cognition allows individuals to rapidly mediate their behaviour toward conspecifics, including approach and avoidance, in a manner that is adaptive within dynamic social contexts [19].
Approach can be defined as the direction of behaviour toward positive stimuli (higher incentive salience and reward), while avoidance can be defined as directing behaviour away from negative stimuli [57]. Approach and avoidance behaviour can be directed toward objects, individuals, and situations. In terms of the infection risk, avoidance can be defined as any host defence that reduces the rate of contact between a potential host and parasite [16]. Pathogen avoidance behaviour can be undertaken through: (i) directly avoiding or removing visible parasites; or (ii) indirectly avoiding individuals or altering interactions with individuals that may be a potential source of parasites and infection [19,56,58]. Once a pathogen threat has been detected, social avoidance serves as the first behavioural defence against infection that an individual mounts and is likely the most effective action to protect against infection [16]. This has been termed the behavioural immune system in human studies [59]. Pathogen avoidance is, however, context-dependent and needs to be balanced against other behaviours that are essential for survival and reproduction. Context-related factors (e.g., food availability, predator exposure, etc.) need to be incorporated into future laboratory studies.
Behavioural avoidance of parasitized and immuno-activated conspecifics has been documented in various rodent species. The odours of healthy conspecifics have been shown to elicit approach behaviours, while the odours of individuals with infection or immune activation are shown to induce avoidance [19,60,61]. For example, treatment of female mice with the endotoxin lipopolysaccharide (LPS) resulted in various behavioural avoidance responses, such as increased inter-individual distance, decreased rates of physical contact, and the modality of social exploration (i.e., decreased anogenital sniffing) [62]. Similarly, male rats demonstrated significant suppression of social interaction directed toward LPS-treated male conspecifics [63]. Additionally, female prairie voles spent more time in close proximity to a healthy male in comparison to an LPS-treated male [64].
Regarding zoonotic parasites specifically, multiple studies have demonstrated that rodents tend to also avoid conspecifics carrying nematode parasites (i.e., helminths) and ectoparasites (i.e., lice and ticks). Female mice avoid the urinary odours of males infected with the louse Polyplax serrata [54]. Edwards (1988) [65] found that healthy male mice engaged in increased olfactory investigation of conspecific males infected with Trichinella spiralis but engaged in less physical contact, suggesting the processing of olfactory cues associated with infection is important in the elicitation of avoidance behaviour. Female rats, mice, and meadow voles have also all been shown to similarly discriminate against males infected with various species of parasitic nematodes [28,66,67].

3. Olfactory Mediated Detection of and Response to Infection Status

Rodents rely heavily on olfaction to provide social information about the status and condition of conspecifics, including that of potential mates and social partners. Chemosignals are informative of an individual’s identity and condition, including infection status [53,68,69,70,71]. The rodent olfactory system is subdivided into two primary components, the main olfactory system (consisting of the main olfactory epithelium (MOE)) and the vomeronasal system (VNO) [72,73,74]. The MOE projects to the main olfactory bulb, while the VNO projects to the accessory olfactory bulb [74]. Rodents emit various odours consisting of volatile (detected directly through the MOE) and non-volatile (detected through the VNO) chemical constituents that serve as an indication of a conspecific’s physiological state [23,75]. In mice, pheromone chemical cues are typically present in urine and convey information about health status [76,77]. However, the roles of other odour sources, particularly that of the preputial gland in males, need to be further examined (for a discussion of odour sources and sexual attraction, see Le Moëne and Ågmo (2018) [78]).
Non-volatile olfactory cues in the bodily products of mice trigger neural activity in the VNO, which is specifically involved in the expression of various innate behaviours [73,79]. Boillat et al. (2015) [73] demonstrated that mice with impaired VNO function did not demonstrate the typical preference toward healthy conspecific versus conspecifics treated with LPS. The researchers verified that the aversive infection cues detected by the VNO were present in bedding and urine specifically, as mice with intact VNO function preferentially interacted with bedding materials from healthy conspecific over those of LPS-treated individuals. It should be noted here that bedding contains odour cues from a range of bodily sources. However, the results of cFos imaging found that exposure to LPS-infected urine resulted in higher neural activity in the accessory olfactory bulb, the region to which the VNO projects.
Although the MOE is not considered to be primarily involved in the detection of pheromones, there is evidence that pheromone-based behaviour is mediated by both the MOE and the main olfactory bulb [74,80,81]. The main olfactory bulb in mice has small populations of mitral cells that respond to volatile urine compounds. Lin et al. (2005) [81] demonstrated that in female mice, subsets of these olfactory mitral cells responded exclusively to male urine due to a novel male-specific pheromone, (methylthio)methanethiol. When added to urine, synthetic (methylthio)methanethiol enhanced female attraction to male urine, suggesting that it may be a volatile compound processed by the MOE and main olfactory bulb that serves in the female assessment of males. Whether or not these constituents are involved in mate choice remains to be determined.
Evidence suggests that urinary odours are involved in conveying information influencing mate choice in rodents. For example, the results of studies have shown that female laboratory mice can discriminate the urinary odours of male mice infected with the helminth Heligmosomoides polygyrus and display a preference for the urinary odours of uninfected males [29,82]. Females may be responding to subtle differences in male urinary odours, likely associated with polymorphic gene complexes: the major histocompatibility complex (MHC) and the major urinary proteins (MUP) [68,83]. These gene complexes convey an array of information about sex, kinship, dominance, age, and health status [23]. Olfactory processing of odours containing MHC in particular serves as an indication of genotype and physiological quality and is used to assess immune function [84,85,86]. MUPs have considerable polymorphic expression between individuals and serve mainly as markers of genetic identity (i.e., kinship, genetic heterozygosity), and they are also implicated in the assessment of immune status [76,87]. Only these urinary compounds were sufficient for the detection of conspecific infection status, as feces and bile acid from mice infected with the endotoxin LPS did not induce avoidance in conspecifics [88]. The detection and processing of volatile urinary components allows individuals to identify the producers of infected urine from a distance and avoid close or direct contact with any potential pathogen products.
Parasitic infection has been shown to affect MUP expression in mice. Li et al. (2023) [76] demonstrated that the urine of male mice infected with Cryptosporidium parvum displayed lower attractiveness to females, and they additionally showed that the protein concentration of MUP in the urine was downregulated in parasitized males. In addition to this, the contents of different pheromones were downregulated in the preputial gland of these males, suggesting that parasitism alters the concentration of male pheromones. An additional study by Li et al. (2023) [77] demonstrated that the parasite Trichinella spiralis had similar effects on male MUP expression and decreased sperm quality.
This short discussion reveals that a range of volatile and non-volatile pheromone attractants are produced by male and female rodents. These chemical factors are genetically controlled and likely influenced by the social and non-social environmental context (e.g., the presence of conspecifics of varying status (dominance) and physiological condition). There is evidence that mouse urinary volatiles vary according to the pathogen responsible for producing inflammation [89]. As well, the extent of avoidance has been linked to the degree of infection [90]. How infection affects these chemical signals, both in terms of quality and quantity, remains to be resolved. Comparative transcriptomics analysis examining changes in gene expression in relation to odorants in infected and non-infected individuals is required to complement the analysis of the constituents.

4. Neurobiological Activation of Affective and Cognitive Responses to the Pathogen Threat

In recent years, the neurobiological mechanisms underlying the assessment of the infection status of conspecifics have gained significant interest [91]. Here, we will discuss this research and briefly consider the brain regions, hormones, and circuits involved in the expression of disgust and pathogen avoidance in rodents.

4.1. Brain Regions

The processing of pathogen detection and the initiation of appropriate responses involve various brain regions, including the evolutionary conserved social decision-making network. This includes areas such as the social brain and the mesocorticolimbic reward network, which are composed of areas such as the medial amygdala, insular cortex, and related regions [92,93,94]. These are briefly considered below (see Figure 2 for a diagram of these regions and a simplified depiction of their projections relevant to pathogen avoidance).

4.1.1. Medial Amygdala

The medial amygdala (MeA) is a critical region for the decoding of social stimuli and processing of appropriate behavioural responses. In rodents, both the main and accessory olfactory bulbs have projections directly to the MeA [95,96,97]. The majority of the olfactory projections to the MeA come from the accessory olfactory bulb (and indirectly from the VNO), suggesting that the MeA is involved in the processing of non-volatile chemical sensory information primarily detected by the accessory olfactory bulb [98,99]. In turn, the MeA is functionally connected to a broad network of limbic regions and the hypothalamus, suggesting that the MeA is involved in the transformation of sensory information into specific behavioural responses [98,100,101].
The MeA is differentially implicated in both appetitive and aversive/avoidance behaviour, with it and its projections mediating appetitive responses to reproductive stimuli, as well as the processing and avoidance of aversive odours [101,102,103]. The integration of olfactory signal and social stimuli processing in the MeA likely makes the region integral in both the detection of, and the behavioural response to, parasitic risk.
In male rats, odour from conspecifics treated with LPS has been shown to induce avoidance response in healthy rats. Arakawa et al. (2010) [104] found that the MeA and multiple brain regions that it projects to were activated (including the bed nucleus of the stria terminalis (BNST) and the paraventricular nucleus of the hypothalamus (PVN)) in rats exposed to the odour of a sick LPS-treated conspecific, exemplifying the role of the MeA as a hub of social information processing. In this regard, whether the responses were elicited by exposure to sickness, which elicits a constellation of responses, or to LPS treatment per se remains to be determined.
Kwon et al. (2021) [105] identified the posteromedial nucleus of the cortical amygdala (COApm) as a region involved in the suppression of mating behaviour in male mice when exposed to an LPS-treated female. Using functional imaging, the COApm was shown to project to populations of glutamatergic neurons in the MeA. Interestingly, Kwon et al. (2021) [105] also demonstrated that the thyrotropin-releasing hormone (TRH) and its receptor within the MeA are implicated in the suppression of mating with LPS-treated females. Male mice with genetically ablated TRH receptors engaged in mating with unhealthy individuals, suggesting that these receptors in the MeA are involved in avoiding interactions and mating with conspecifics displaying cues of enhanced immune activity.
A recent study investigating the Gai2 vomeronasal system in mouse sickness avoidance behaviour found that the Gai2 pathway is required for the detection and avoidance of LPS-treated conspecific urine [87]. Gai2-dependent neuronal activity occurred in the MeApv when exposed to the urine of LPS-treated conspecifics, suggesting that the region is involved in the detection and avoidance of sick conspecifics. Interestingly, the neuronal activation was not as strong in mice exposed to LPS-treated urine compared to the neuronal activation of control mice exposed to non-infected urine. This may be indicative of the region’s role in social processing and motivation, with the lower MeApv activity in response to LPS-urine potentially contributing to the lack of approach behaviour.
Together, these findings indicate that the MeA serves as one of the first processing points of conspecific chemosignals, facilitating the detection of pathogenic threats and eliciting subsequent behavioural responses, such as approach and mate choice. As such, the MeA is crucial to the detection of social signals that are indicative of pathogenic threats and likely plays a key role in triggering behavioural avoidance and disgust responses through its projections to other brain regions. Which specific brain regions, and particularly how these may be integrated to facilitate behavioural responses, remains to be determined.

4.1.2. Insular Cortex

The insular cortex is a region heavily implicated in the disgust response and the mediation of various aversive/avoidance behaviours through connections with both social brain regions and reward regions [91,106,107,108]. The insular cortex can be divided into two distinct regions: the posterior insular cortex and the anterior insular cortex. The posterior insular cortex projects to the sensory and limbic brain regions, while the anterior insular cortex mainly projects to the social brain network, including the medial prefrontal cortex, the nucleus accumbens, and the amygdala [108,109].
The insular cortex is directly involved in pathogen avoidance, with inactivation of the posterior insular cortex eliminating the avoidance response to conspecifics treated with a viral mimetic in both female and male rats [110]. A recent study by Min et al. (2023) [109] demonstrated that lesions on the anterior insular cortex of male mice resulted in an inability to distinguish between a cage mate and a novel mouse. Due to the role of the anterior insular cortex in processing information leading to social recognition, this region may also be involved in the processing of infection cues and the recognition, and avoidance, of parasitized individuals.
A study investigating the correlation between the stereotyped facial expression and the affective state in mice found that exposure to a toxin (lithium chloride) elicited facial expressions that were associated with disgust and corresponded to increased activity in the anterior insular cortex [111]. It should be noted that to date, there are no reports that exposure to the odours of infected individuals elicits disgust-like facial expressions in rodents. More detailed machine-learning assessments of facial responses to infection cues are needed.
In mice, the insular cortex also receives monosynaptic input from the MeA, with this anatomical connection being considered to be important in valence processing and emotional regulation [109,112,113]. Additionally, the expression of a disgust-like response to toxins in mice involves neural pathways from the anterior insula to the medial and basolateral amygdala [114]. These neural projections between the insular cortex and the MeA denote a functional link between the two regions in the processing of aversive social stimuli and disgust-like responses.

4.1.3. Habenula

The habenula is a highly conserved limbic brain region implicated in divergent motivational states and cognition [115]. Subnuclei of the habenula are thought to be involved in the processing of avoidance responses to various stimuli. The lateral habenula is central to the brain’s reward system and inhibits dopamine release in the midbrain to signal negative valence [116]. Studies using fibre-photometry found that the lateral habenula of mice shows increased neuronal activity when the mouse was presented with a bitter taste, pain, and aggression from a conspecific [117]. In addition to this, the lateral habenula is also involved in the regulation of social behaviour, with chemogenetic activation of the region resulting in a reduction in social behaviour and social interactions [93,118]. Wang et al. (2017) [117] suggested that aversion processing in the lateral habenula is highly influenced by learning, as a prolonged positive reward inhibited lateral habenula activity in mice that were conditioned to an aversive stimulus. This suggests that lateral habenula activity in response to, and processing of, aversive stimuli is experience dependent.
Weiss et al. (2023) [87] further implicated the lateral habenula in the detection and avoidance of the urine of LPS-treated conspecifics. Exposure to LPS-treated conspecific urine resulted in lateral habenula activation four times stronger than the activity seen during exposure to control urine. Although the habenula is often described as the region associated with hedonia and anhedonia, more consideration should be focused on this region when studying disgust and aversion to pathogens, as positive and negative affect likely heavily influence adaptive responses to stimuli that evoke disgust.

4.1.4. Nucleus Accumbens

Recent research has explored the role of the nucleus accumbens (NAc) in the avoidance of aversive stimuli. The NAc is integral in the detection of aversive stimuli and translating motivation into action. The NAc is a convergence point of both reward and aversion circuitry, receiving projections from the ventral tegmental area, the medial prefrontal cortex, and the basolateral amygdala [119,120,121,122]. Specifically, dopamine signalling from the ventral tegmental area has been linked to reward and aversion processing in the NAc, while glutamatergic signalling from the thalamic paraventricular nucleus to the NAc is implicated in aversion [123,124]. He et al. (2023) [119] found that Tac1 neurons in the medial shell of the NAc are specifically implicated in the mediation of the avoidance response to aversive olfactory stimuli (formaldehyde) by receiving excitatory glutamatergic input from the medial prefrontal cortex and sending inhibitory signals to the lateral hypothalamic area.
The NAc is also involved in social reward, sexual behaviour, and mating [125,126,127]. In both male and female mice, approach and investigation of an opposite sex conspecific result in dopamine activation in the NAc, while dopamine signalling in the NAc is sexually dimorphic during sexual behaviours [128]. Additionally, the role of the neuropeptide oxytocin (which is highly implicated in social behaviour) in the NAc has been shown to regulate social preference and other aspects of social behaviour related to sexual behaviour [129,130,131].
Given the role of the NAc in both sexual behaviour and approach/avoidance, it is likely that it is an important region in mate assessment and avoidance of parasitic risk. The NAc and other regions of the mesolimbic reward network are associated with the determination of the incentive salience and reward value of a potential mate [38,132,133]. As such, how the cues of infected individuals influence the functioning of the reward network components (specifically dopamine levels) and subsequent approach/avoidance regarding mate choice remains to be determined.

4.1.5. Concluding Statement on Brain Regions

This short discussion summarizes the brain regions and their interconnections (circuits) shown to be involved in the mediation of responses to infection threats in rodents. Whether these regions and circuits are similarly involved in the initiation and maintenance of avoidance responses remains to be determined. In addition, a range of other brain regions, including those in the social brain network (e.g., medial prefrontal cortex, basal ganglia, ventral tegmental area), are implicated in the processing of defensive responses to a variety of threats, likely including those to the pathogen threat (for a discussion, see Tseng et al., 2023 [134]). Notably, the prefrontal cortex has been recently shown to encode both general and specific threat representations [135]. In this regard, studies with humans showed that pictures of sick LPS-treated individuals led to decreased activity in the ventromedial prefrontal cortex and the initiation of avoidance [136].

4.1.6. Immune–Brain Interactions

The roles of peripheral systems (e.g., immune systems) and their interactions with central systems in the modulation of the behavioural avoidance of the pathogen threat need consideration. Infection- and disease-associated contexts have been shown to trigger immune responses [63,137,138]. It is likely that social cues of infection can alter immune responses in uninfected individuals. Indeed, recent work has suggested a co-evolution of social behaviour and immune responses. A zinc finger transcription factor, ZFP189, was shown to modulate social behaviour by controlling transposable elements and immune responses in the prefrontal cortex of mice [139]. This was based on earlier work suggesting a co-evolutionary link between social behaviour and anti-pathogen immune responses driven by interferon-γ signalling [140]. ZFP182 upregulated immune genes that are involved in anti-pathogen responses, while also promoting social behaviour. Moreover, these responses were suggested to be responsive to environmental stressors, including possibly that of the parasite/infection threat. It was further speculated that other transcription factors may regulate social behaviour and immune responses. These findings provide an exciting new direction for the elucidation of the neurobiological regulation of pathogen avoidance.

4.1.7. Microbiome–Immune–Brain Interactions

Animals have diverse populations of bacteria and viruses both internally and externally [141,142,143,144]. There is now substantial evidence that the gastrointestinal microbiota (microbiome) can signal to the brain (microbiota–gut–brain axis) through a range of pathways, including immune activation; production of microbial metabolites and peptides; alterations in neurotransmitters and hormonal levels; neuromodulators and activation of the vagus nerve [145]. These actions can influence various brain regions, including those of the social brain, with marked implications for pathogen avoidance and mate choice.
The microbiome plays a role in influencing the immune response and, subsequently, social behaviour and pathogen avoidance. The microbiome can influence social behaviour through the production of, and effect on, chemosignals [141,146,147]. Microbial metabolism can release volatile compounds that are detectable by conspecifics and may influence olfactory social signalling [148]. Studies in fruit flies (Drosophila melanogaster) have shown that the gut microbiota composition mediates olfactory-based mate preference [149]. More pertinent to this review, Li et al. (2013) [150] demonstrated that trimethylamine (TMA), which is a volatile compound involved in the signalling of species identity, is influenced by the microbiota in mice. TMA is produced by gut bacteria during choline metabolism and is converted into an ‘odorless’ compound by enzymes in the liver. However, these liver enzymes are downregulated in sexually mature male mice, allowing TMA and its odour to be detectable in the urine. When male mice were administered antibiotics, they produced significant quantities of TMA, suggesting the direct implication of the microbiome in producing this chemosignal.
Additional studies have shown that the sexual attractiveness of female mice may be impacted by dysbiosis of the gut microbiome [151]. Yi and Cha (2022) [151] found that male mice showed more interaction with control females in a three-chamber social test in comparison to females that had a gut microbiome manipulated by antibiotic treatment. The authors suggested that the decrease in sexual attractiveness in antibiotic-treated females may be due to alterations to the immune system [151]. It is also possible that dysbiosis of females may be influencing the chemosignals through hormonal effects as well. Some research has coined the term ‘Estrogen–Gut Microbiome Axis’, highlighting the influence that the microbiome has on the estrogenic system [152,153]. Multiple studies have shown that alterations to the circulating estrogen and androgen levels impact gut microbiota composition [154,155,156,157,158,159]. Given that estrogens are heavily implicated in olfactory and pheromonal cues, it is likely that reduced female attraction caused by dysbiosis may be a result of altered estrogen levels. Altered estrogen levels may influence the production of other pheromones and ultimately influence the attractiveness of the female [160,161].
Additionally, some interesting research has directly investigated the influence of parasitism on the microbiome. These interactions are complex and are mediated by the environment, genetics, and the infection burden [162]. Zhao et al. (2019) [163] demonstrated that female mice infected with the helminth parasite Schistosoma japonicum had a significant influence on the gut microbiome composition, likely caused by the parasitic egg granulomas in the intestinal tissue. Preliminary data show that the gut microbiome composition is affected by tick parasitism in mice [164]. A number of additional studies in rodents have shown an interaction between parasitic infection and the host microbiome [163,165,166,167].
To date, much of the work on the effect of the microbiome composition and how it influences mate choice has been conducted in non-rodent species [149,168,169]. This leaves an exciting avenue for rodent researchers to investigate and establish how the microbiome composition (including both bacterial and viral elements), and the impact of parasitism on this composition, may influence mate choice and social behaviour. Integral to this is also determining how the microbiome influences the expression of affective states such as disgust, fear, and anxiety.

4.2. Neuropeptides and Hormones Involved in Pathogen Avoidance

Many of the brain regions, and likely the microbiome components, considered above involve the actions of various neuromodulatory systems: various neuropeptide systems, neurotransmitter systems, sex steroid hormones and other steroids. These are briefly considered below.

4.2.1. Oxytocin

The neuropeptide oxytocin is implicated in various aspects of social behaviour, such as the processing of social information and social memory in mice and various other rodent species [170,171]. In addition to mediating social recognition and peer affiliation, oxytocin is involved in the olfactory-mediated recognition, and avoidance of, infected individuals. In rodents, oxytocin receptors modulate the ‘social salience network’, a collection of interconnected brain nuclei involved in the processing and encoding of social and sensory cues and involved in the processing of cues associated with the expression of disgust [171,172,173,174].
Female mice with both genetic and pharmacological impairment to oxytocin receptor function displayed decreased aversion and increased choice toward parasitized male conspecific odour compared to control females in a mate-choice paradigm [175]. Additionally in female mice, the preference for parasitized and non-parasitized conspecific male odours was affected by the administration of an oxytocin receptor antagonist, suggesting that oxytocin is involved in the rapid decision-making associated with the approach or avoidance of a parasitic threat [48]. These findings suggest that oxytocin may play a central role in the recognition and subsequent avoidance of parasitized males by females.
A study on conditioned disgust responses in male rats found that oxytocin modulates the expression of socially mediated disgust. When male rats with lithium chloride (LiCl)-induced sickness were conditioned with a social partner, they showed an increased mouth-gaping response (indicative of disgust and malaise) when re-exposed to the conditioned partner in comparison to exposure to a novel conspecific. Boulet et al. (2016) [176] demonstrated that when given an oxytocin receptor antagonist, rats that were conditioned with LiCl showed significantly less disgust (mouth gaping) when re-exposed to the conditioned partner, suggesting that functional oxytocin receptors are required to develop an associative disgust response toward a conspecific. As such, oxytocin may modulate both disgust-associated social salience and social recognition, integral components of mate choice.
What oxytocin-associated pathways are involved in mediating these effects remains to be determined. Also, whether these modulatory roles arise from the effects of oxytocin on other neurotransmitter/neuromodulatory pathways remains to be determined. In this regard, oxytocin has interactions with estrogens and dopamine, and likely immune components, in the regulation of social behaviours [177]. Both dopaminergic and estrogenic systems are implicated in the regulation of female mate choice and, as such, pathogen avoidance.

4.2.2. Vasopressin

The neuropeptide vasopressin has also been shown to influence the avoidance of pathogen risk. Arakawa et al. (2010) [104] demonstrated that vasopressin within the MeA of rats is involved in conspecific infection avoidance. When exposed to the odours of LPS-treated male conspecifics, the expression of vasopressin messenger RNA in the MeA increased. Infusion of the vasopressin receptor (V1A) antagonist to the MeA inhibited the previously demonstrated avoidance response to LPS-treated odours.
Vasopressin is structurally very similar to oxytocin and has a high homology for oxytocin receptors as well as vasopressin (AVP) receptors. Due to this homology, the vasopressin system may likely interact with, and influence, the oxytocin system and the role that it is known to play in pathogen and sickness detection and avoidance. Similarly, as for oxytocin, the roles of other neurotransmitter/neuromodulatory systems remain to be examined regarding the potential interactions with vasopressin.

4.2.3. Estrogens

Estrogens are a class of steroid hormones that affect multiple physiological functions and behaviours in both males and females. In females, heightened sexual motivation is mainly influenced by estrogens, which in turn influence dopamine signalling in the mesolimbic reward centres of the brain [59,178,179]. The salience of a potential mate’s cues can influence the sexual preference and decision made. Lynch and Ryan (2020) [59] postulated that estrogens increase female interest in mating, while the estrogenic influence on dopamine facilitates the detection of signals with high incentive versus low incentive and informs mate preference.
There is also evidence that estrogenic mechanisms are involved in the processing and avoidance of conspecific infection cues [175,180,181]. Kavaliers et al. (2004) [175] showed that female ERα and ERβ receptor knock-out mice had impairments in the discrimination and avoidance of urine odour in both males that were either treated with LPS or infected with a nematode parasite. This suggests that estrogenic mechanisms are involved in the recognition of, and response to, sick and infected conspecifics. Male ERα and ERβ receptor knock-out mice showed similar impairment in their response to the urine odours of sick and infected males [71]. The inability of ERα and ERβ receptor knock-out mice to recognize cues of infection was shown to not be the result of impaired olfactory processing, as other olfactory-based paradigms were not impaired (such as the ability to distinguish the sexual status of conspecifics and aversion to predator odour).
Additionally, all three estrogen receptors (ERα, ERβ, GPER) are expressed in the MeA, where they are involved in enhancing social recognition and interact with oxytocin receptors, suggesting that estrogens are implicated in the MeA’s role in the detection of and behavioural response to parasitized conspecifics [180,182,183].
Given the roles that estrogenic mechanisms play in olfactory-mediated social recognition, response to pathogenic threat, influence on immune function, and sexual motivation/behaviour, estrogens are integral to the processing of salient social information that informs the adaptive behavioural response to pathogenic risk [180,184,185,186,187]. More research investigating how estrogenic mechanisms interact and potentially link these physiological, cognitive, and behavioural processes is required.

4.2.4. Progesterone

In human research, the compensatory prophylaxis hypothesis states that the luteal phase of the menstrual cycle (characterized by high progesterone levels) suppresses various mechanisms of immune response and thus should result in a heightened disgust response (reviewed in Fleischman and Fessler, 2011 [188]). Recent evidence suggests that progesterone plays a part in the rodent disgust response and pathogen avoidance. In a re-analysis of the results of Kavaliers et al. (2021) [189] conducted by Bressan and Kramer (2022) [190], estrous female mice administered exogenous progesterone had an enhanced avoidance response to the odours of a nematode-infected male.
The biological mechanisms that progesterone may be acting on to influence a disgust-like response remain to be determined. However, progesterone may influence sensory inputs involved in detecting salient social information, as progesterone was shown to potentially influence social recognition [189]. More research into the potentially enhancing effects of progesterone and its neurosteroid products on pathogen detection and disgust-like response needs to be conducted before further speculation.

4.2.5. Concluding Statement on Neuropeptides and Hormones

We have highlighted the neuromodulatory mechanisms that have been directly implicated in the modulation of pathogen detection and avoidance as they relate to mate choice. There are a range of other regulatory systems that need consideration (e.g., immune system components, opioid systems, neurotransmitters (e.g., 5-HT, endocannabinoids) and steroid and non-steroid hormones). For example, there is evidence that avoidance of infection risk is driven by physiological responses associated with stress and anxiety. Lopes (2023) [191] speculated that after the initial detection of parasite risk, the sympathetic nervous system, adrenomedullary response and adrenocortical response are triggered to facilitate avoidance. This reinforces the need to further consider the roles of, and links between, disgust, fear, and anxiety in modulating pathogen detection and avoidance.

5. Conclusions

The main objective of this review was to draw attention to numerous issues of basic importance for understanding the neurobiology of pathogen avoidance and mate choice in rodents. We considered avoidance behaviours concerning potential and actual pathogenic threats, the nature of the chemical stimuli eliciting these responses, and some of the mechanisms determining and controlling responsiveness. Due to the high fitness cost of parasitism, there is strong evolutionary pressure to develop efficient systems for parasite detection and avoidance [22]. Avoidance of parasitized conspecifics is a highly conserved evolutionary trait and is universal throughout the animal kingdom [18,36,192]. To date, the majority of studies examining the neurobiology of pathogen avoidance and its relation to mate choice have been limited to laboratory conditions and have primarily considered female responses. The roles of the environmental context (e.g., presence of predators, threatening conspecifics), trade-offs with other factors (food and mate availability, own infection status), and uncertainty (what is the likelihood of the conspecific still being infectious) all need to be considered and incorporated in future studies.

Author Contributions

D.C.; writing-original draft preparation, M.K.; writing-review and editing, E.C.; supervision. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by NSERC grants to E.C. (grant number RGPIN-2018-04699) and M.K. (grant number RGPIN-2022-03886).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The research and data discussed in this review are cited and are available in peer-reviewed journals.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Schmid-Hempel, P. Parasites and Their Social Hosts. Trends Parasitol. 2017, 33, 453–462. [Google Scholar] [CrossRef] [PubMed]
  2. Favreau, F.-R.; Goldizen, A.W.; Pays, O. Interactions among social monitoring, anti-predator vigilance and group size in eastern grey kangaroos. Proc. R. Soc. B Biol. Sci. 2010, 277, 2089–2095. [Google Scholar] [CrossRef] [PubMed]
  3. Gersick, A.S.; Snyder-Mackler, N.; White, D.J. Ontogeny of social skills: Social complexity improves mating and competitive strategies in male brown-headed cowbirds. Anim. Behav. 2012, 83, 1171–1177. [Google Scholar] [CrossRef]
  4. Nöbel, S.; Wang, X.; Talvard, L.; Tariel, J.; Lille, M.; Cucherousset, J.; Roussigné, M.; Danchin, E. The importance of population heterogeneities in detecting social learning as the foundation of animal cultural transmission. Proc. R. Soc. B Biol. Sci. 2022, 289, 20220431. [Google Scholar] [CrossRef]
  5. Hofmann, H.A.; Beery, A.K.; Blumstein, D.T.; Couzin, I.D.; Earley, R.L.; Hayes, L.D.; Hurd, P.L.; Lacey, E.A.; Phelps, S.M.; Solomon, N.G.; et al. An evolutionary framework for studying mechanisms of social behavior. Trends Ecol. Evol. 2014, 29, 581–589. [Google Scholar] [CrossRef]
  6. Sah, P.; Mann, J.; Bansal, S.; Farine, D. Disease implications of animal social network structure: A synthesis across social systems. J. Anim. Ecol. 2018, 87, 546–558. [Google Scholar] [CrossRef] [PubMed]
  7. Rifkin, J.L.; Nunn, C.L.; Garamszegi, L.Z. Do Animals Living in Larger Groups Experience Greater Parasitism? A Meta-Analysis. Am. Nat. 2012, 180, 70–82. [Google Scholar] [CrossRef]
  8. Patterson, J.E.H.; Ruckstuhl, K.E. Parasite infection and host group size: A meta-analytical review. Parasitology 2013, 140, 803–813. [Google Scholar] [CrossRef]
  9. Altizer, S.; Harvell, D.; Friedle, E. Rapid evolutionary dynamics and disease threats to biodiversity. Trends Ecol. Evol. 2003, 18, 589–596. [Google Scholar] [CrossRef]
  10. Kappeler, P.M.; Cremer, S.; Nunn, C.L. Sociality and health: Impacts of sociality on disease susceptibility and transmission in animal and human societies. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2015, 370, 20140116. [Google Scholar] [CrossRef]
  11. Freeland, W.J. Pathogens and the Evolution of Primate Sociality. Biotropica 1976, 8, 12–24. [Google Scholar] [CrossRef]
  12. Ezenwa, V.O.; Ghai, R.R.; McKay, A.F.; Williams, A.E. Group living and pathogen infection revisited. Curr. Opin. Behav. Sci. 2016, 12, 66–72. [Google Scholar] [CrossRef]
  13. Elton, C.; Ford, E.B.; Baker, J.R.; Gardner, A.D. The Health and Parasites of a Wild Mouse Population. Proc. Zool. Soc. Lond. 1931, 101, 657–721. [Google Scholar] [CrossRef]
  14. Gregory, R.D.; Montgomery, S.S.J.; Montgomery, W.I. Population Biology of Heligmosomoides polygyrus (Nematoda) in the Wood Mouse. J. Anim. Ecol. 1992, 61, 749–757. [Google Scholar] [CrossRef]
  15. Iwanowicz, D.D. Overview on the effects of parasites on fish health. In Bridging America and Russia with shared Perspectives on Aquatic Animal Health: Proceedings of the Third Bilateral Conference between Russia and the United States, Sheperdstown, WV, USA, 12 July 2009; Khaled bin Sultan Living Oceans Foundation: Landover, MD, USA, 2011; pp. 176–184. [Google Scholar]
  16. Gibson, A.K.; Amoroso, C.R. Evolution and Ecology of Parasite Avoidance. Annu. Rev. Ecol. Evol. Syst. 2022, 53, 47–67. [Google Scholar] [CrossRef] [PubMed]
  17. Curtis, V.; Biran, A. Dirt, Disgust, and Disease: Is Hygiene in Our Genes? Perspect. Biol. Med. 2001, 44, 17–31. [Google Scholar] [CrossRef]
  18. Curtis, V. Why disgust matters. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2011, 366, 3478–3490. [Google Scholar] [CrossRef]
  19. Kavaliers, M.; Choleris, E. The role of social cognition in parasite and pathogen avoidance. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2018, 373, 20170206. [Google Scholar] [CrossRef]
  20. Sarabian, C.; Wilkinson, A.; Sigaud, M.; Kano, F.; Tobajas, J.; Darmaillacq, A.-S.; Kalema-Zikusoka, G.; Plotnik, J.M.; MacIntosh, A.J.J. Disgust in animals and the application of disease avoidance to wildlife management and conservation. J. Anim. Ecol. 2023, 92, 1489–1508. [Google Scholar] [CrossRef]
  21. van Leeuwen, F.; Jaeger, B.; Tybur, J.M. A behavioural immune system perspective on disgust and social prejudice. Nat. Rev. Psychol. 2023, 2, 676–687. [Google Scholar] [CrossRef]
  22. Weinstein, S.B.; Buck, J.C.; Young, H.S. A landscape of disgust. Science 2018, 359, 1213–1214. [Google Scholar] [CrossRef]
  23. MacLellan, A.; Adcock, A.; Mason, G. Behavioral biology of mice. In Behavioral Biology of Laboratory Animals; CRC Press: Boca Raton, FL, USA, 2021; pp. 89–111. [Google Scholar]
  24. Dass, S.A.H.; Vasudevan, A.; Dutta, D.; Soh, L.J.T.; Sapolsky, R.M.; Vyas, A. Protozoan parasite Toxoplasma gondii manipulates mate choice in rats by enhancing attractiveness of males. PLoS ONE 2011, 6, e27229. [Google Scholar] [CrossRef]
  25. Ehman, K.D.; Scott, M.E. Urinary odour preferences of MHC congenic female mice, Mus domesticus: Implications for kin recognition and detection of parasitized males. Anim. Behav. 2001, 62, 781–789. [Google Scholar] [CrossRef]
  26. Kavaliers, M.; Fudge, M.A.; Colwell, D.D.; Choleris, E. Aversive and Avoidance Responses of Female Mice to the Odors of Males Infected with an Ectoparasite and the Effects of Prior Familiarity. Behav. Ecol. Sociobiol. 2003, 54, 423–430. [Google Scholar] [CrossRef]
  27. Penn, D.; Schneider, G.; White, K.; Slev, P.; Potts, W. Influenza Infection Neutralizes the Attractiveness of Male Odour to Female Mice (Mus musculus). Ethology 1998, 104, 685–694. [Google Scholar] [CrossRef]
  28. Willis, C.; Poulin, R. Preference of female rats for the odours of non-parasitised males: The smell of good genes? Folia Parasitol. 2000, 47, 6–10. [Google Scholar] [CrossRef]
  29. Kavaliers, M.; Colwell, D.D. Odours of parasitized males induce aversive responses in female mice. Anim. Behav. 1995, 50, 1161–1169. [Google Scholar] [CrossRef]
  30. Able, D.J. The Contagion Indicator Hypothesis for Parasite-Mediated Sexual Selection. Proc. Natl. Acad. Sci. USA 1996, 93, 2229–2233. [Google Scholar] [CrossRef]
  31. Kokko, H.; Brooks, R.; Jennions, M.D.; Morley, J. The evolution of mate choice and mating biases. Proc. R. Soc. B Biol. Sci. 2003, 270, 653–664. [Google Scholar] [CrossRef]
  32. Loehle, C. Social Barriers to Pathogen Transmission in Wild Animal Populations. Ecology 1995, 76, 326–335. [Google Scholar] [CrossRef]
  33. Raveh, S.; Sutalo, S.; Thonhauser, K.E.; Thoß, M.; Hettyey, A.; Winkelser, F.; Penn, D.J. Female partner preferences enhance offspring ability to survive an infection. BMC Evol. Biol. 2014, 14, 14. [Google Scholar] [CrossRef] [PubMed]
  34. Hashikawa, K.; Hashikawa, Y.; Falkner, A.; Lin, D. The neural circuits of mating and fighting in male mice. Curr. Opin. Neurobiol. 2016, 38, 27–37. [Google Scholar] [CrossRef]
  35. Beny-Shefer, Y.; Zilkha, N.; Lavi-Avnon, Y.; Bezalel, N.; Rogachev, I.; Brandis, A.; Dayan, M.; Kimchi, T. Nucleus Accumbens Dopamine Signaling Regulates Sexual Preference for Females in Male Mice. Cell Rep. 2017, 21, 3079–3088. [Google Scholar] [CrossRef] [PubMed]
  36. Kavaliers, M.; Wah, D.T.O.; Bishnoi, I.R.; Ossenkopp, K.-P.; Choleris, E. Disgusted snails, oxytocin, and the avoidance of infection threat. Horm. Behav. 2023, 155, 105424. [Google Scholar] [CrossRef]
  37. Ventura-Aquino, E.; Portillo, W.; Paredes, R.G. Sexual Motivation: A Comparative Approach in Vertebrate Species. Curr. Sex. Health Rep. 2018, 10, 114–123. [Google Scholar] [CrossRef]
  38. Ågmo, A. Sexual motivation—An inquiry into events determining the occurrence of sexual behavior. Behav. Brain Res. 1999, 105, 129–150. [Google Scholar] [CrossRef] [PubMed]
  39. Feagles, O.S.; Höbel, G. Female gray treefrogs maintain mate choice decisions under predation threat, but adjust movements to reduce conspicuousness during mate approach. Behav. Ecol. Sociobiol. 2022, 76, 17. [Google Scholar] [CrossRef]
  40. Beltran-Bech, S.; Richard, F.-J. Impact of infection on mate choice. Anim. Behav. 2014, 90, 159–170. [Google Scholar] [CrossRef]
  41. Dougherty, L.R. Designing mate choice experiments. Biol. Rev. Camb. Philos. Soc. 2020, 95, 759–781. [Google Scholar] [CrossRef]
  42. Dougherty, L.R. The effect of individual state on the strength of mate choice in females and males. Behav. Ecol. 2023, 34, 197–209. [Google Scholar] [CrossRef]
  43. Chu, X.; Guarraci, F.A.; Ågmo, A. Sociosexual behaviors and reproductive success of rats (Rattus norvegicus) in a seminatural environment. Physiol. Behav. 2015, 151, 46–54. [Google Scholar] [CrossRef] [PubMed]
  44. Kavaliers, M.; Choleris, E.; Ågmo, A.; Braun, W.J.; Colwell, D.D.; Muglia, L.J.; Ogawa, S.; Pfaff, D.W. Inadvertent Social Information and the Avoidance of Parasitized Male Mice: A Role for Oxytocin. Proc. Natl. Acad. Sci. USA 2006, 103, 4293–4298. [Google Scholar] [CrossRef] [PubMed]
  45. Galef, B.G., Jr.; Lim, T.C.; Gilbert, G.S. Evidence of mate choice copying in Norway rats, Rattus norvegicus. Anim. Behav. 2008, 75, 1117–1123. [Google Scholar] [CrossRef]
  46. Davies, A.D.; Lewis, Z.; Dougherty, L.R. A meta-analysis of factors influencing the strength of mate-choice copying in animals. Behav. Ecol. 2020, 31, 1279–1290. [Google Scholar] [CrossRef]
  47. Kavaliers, M.; Matta, R.; Choleris, E. Mate-choice copying, social information processing, and the roles of oxytocin. Neurosci. Biobehav. Rev. 2017, 72, 232–242. [Google Scholar] [CrossRef] [PubMed]
  48. Kavaliers, M.; Colwell, D.D.; Wah, D.T.; Bishnoi, I.R.; Ossenkopp, K.P.; Choleris, E. Conspecific infection threat rapidly biases the social responses of female mice: Involvement of oxytocin. Horm. Behav. 2019, 113, 67–75. [Google Scholar] [CrossRef] [PubMed]
  49. Andersson, M. Sexual Selection; Princeton University Press: Princeton, NJ, USA, 1994; Volume 72. [Google Scholar]
  50. Jennions, M.D.; Petrie, M. Variation in mate choice and mating preferences: A review of causes and consequences. Biol. Rev. 1997, 72, 283–327. [Google Scholar] [CrossRef]
  51. Ehman, K.D.; Scott, M.E. Female mice mate preferentially with non-parasitized males. Parasitology 2002, 125, 461–466. [Google Scholar] [CrossRef]
  52. Clayton, D.H. Mate Choice in Experimentally Parasitized Rock Doves: Lousy Males Lose. Am. Zool. 1990, 30, 251–262. [Google Scholar] [CrossRef]
  53. Kavaliers, M.; Colwell, D.D. Discrimination by Female Mice between the Odours of Parasitized and Non-Parasitized Males. Proc. R. Soc. B Biol. Sci. 1995, 261, 31–35. [Google Scholar] [CrossRef]
  54. Kavaliers, M.; Colwell, D.D.; Braun, W.J.; Choleris, E. Brief exposure to the odour of a parasitized male alters the subsequent mate odour responses of female mice. Anim. Behav. 2003, 65, 59–68. [Google Scholar] [CrossRef]
  55. Hamilton, W.; Zuk, M. Heritable true fitness and bright birds: A role for parasites? Science 1982, 218, 384–387. [Google Scholar] [CrossRef]
  56. Kavaliers, M.; Choleris, E.; Pfaff, D.W. Genes, odours and the recognition of parasitized individuals by rodents. Trends Parasitol. 2005, 21, 423–429. [Google Scholar] [CrossRef] [PubMed]
  57. Elliot, A.J. Handbook of Approach and Avoidance Motivation, 1st ed.; Psychology Press: London, UK, 2008. [Google Scholar] [CrossRef]
  58. Hart, B.L. Behavioral adaptations to pathogens and parasites: Five strategies. Neurosci. Biobehav. Rev. 1990, 14, 273–294. [Google Scholar] [CrossRef] [PubMed]
  59. Lynch, K.S.; Ryan, M.J. Understanding the Role of Incentive Salience in Sexual Decision-Making. Integr. Comp. Biol. 2020, 60, 712–721. [Google Scholar] [CrossRef] [PubMed]
  60. Arakawa, H.; Arakawa, K.; Deak, T. Acute Illness Induces the Release of Aversive Odor Cues From Adult, but Not Prepubertal, Male Rats and Suppresses Social Investigation by Conspecifics. Behav. Neurosci. 2009, 123, 964–978. [Google Scholar] [CrossRef] [PubMed]
  61. Arakawa, H.; Arakawa, K.; Deak, T. Sickness-related odor communication signals as determinants of social behavior in rat: A role for inflammatory processes. Horm. Behav. 2010, 57, 330–341. [Google Scholar] [CrossRef]
  62. Renault, J.; Gheusi, G.; Aubert, A. Changes in social exploration of a lipopolysaccharides-treated conspecific in mice: Role of environmental cues. Brain Behav. Immun. 2008, 22, 1201–1207. [Google Scholar] [CrossRef]
  63. Hamasato, E.K.; Lovelock, D.; Palermo-Neto, J.; Deak, T. Assessment of social behavior directed toward sick partners and its relation to central cytokine expression in rats. Physiol. Behav. 2017, 182, 128–136. [Google Scholar] [CrossRef]
  64. Klein, S.L.; Nelson, R.J. Activation of the Immune-Endocrine System With Lipopolysaccharide Reduces Affiliative Behaviors in Voles. Behav. Neurosci. 1999, 113, 1042–1048. [Google Scholar] [CrossRef]
  65. Edwards, J.C. The effects of Trichinella spiralis infection on social interactions in mixed groups of infected and uninfected male mice. Anim. Behav. 1988, 36, 529–540. [Google Scholar] [CrossRef]
  66. Morales, J.; Larralde, C.; Arteaga, M.; Govezensky, T.; Romano, M.C.; Moralí, G. Inhibition of Sexual Behavior in Male Mice Infected with Taenia crassiceps Cysticerci. J. Parasitol. 1996, 82, 689–693. [Google Scholar] [CrossRef] [PubMed]
  67. Klein, S.; Gamble, H.; Nelson, R. Trichinella spiralis infection in voles alters female odor preference but not partner preference. Behav. Ecol. Sociobiol. 1999, 45, 323–329. [Google Scholar] [CrossRef]
  68. Beauchamp, G.K.; Yamazaki, K. Chemical signalling in mice. Biochem. Soc. Trans. 2003, 31 Pt 1, 147–151. [Google Scholar] [CrossRef]
  69. Ferkin, M.H. The response of rodents to scent marks: Four broad hypotheses. Horm. Behav. 2015, 68, 43–52. [Google Scholar] [CrossRef] [PubMed]
  70. Ferkin, M.H. Odor Communication and Mate Choice in Rodents. Biology 2018, 7, 13. [Google Scholar] [CrossRef]
  71. Johnston, R.E. Chemical communication in rodents: From pheromones to individual recognition. J. Mammal 2003, 84, 1141–1162. [Google Scholar] [CrossRef]
  72. Liberles, S.D. Mammalian pheromones. Annu. Rev. Physiol. 2014, 76, 151–175. [Google Scholar] [CrossRef]
  73. Boillat, M.; Challet, L.; Rossier, D.; Kan, C.; Carleton, A.; Rodriguez, I. The Vomeronasal System Mediates Sick Conspecific Avoidance. Curr. Biol. 2015, 25, 251–255. [Google Scholar] [CrossRef]
  74. Wang, Z.; Nudelman, A.; Storm, D.R. Are pheromones detected through the main olfactory epithelium? Mol. Neurobiol. 2007, 35, 317–323. [Google Scholar] [CrossRef]
  75. Stowers, L.; Kuo, T.-H. Mammalian pheromones: Emerging properties and mechanisms of detection. Curr. Opin. Neurobiol. 2015, 34, 103–109. [Google Scholar] [CrossRef] [PubMed]
  76. Li, G.; Zhang, T.; Hu, B.; Han, S.; Xiang, C.; Yuan, G.; He, H. Infection with Cryptosporidium parvum Affects Secondary Sexual Characteristics of Male Mice by Altering the Pheromone Content in Preputial Gland. Animals 2023, 13, 756. [Google Scholar] [CrossRef]
  77. Li, G.; Zhang, T.; Hu, B.; Han, S.; Xiang, C.; Yuan, G.; He, H. Infection of Trichinella spiralis Affects the Reproductive Capacity of ICR/CD-1 Male Mice by Reducing the Urine Pheromone Contents and Sperm Quality. Int. J. Mol. Sci. 2023, 24, 5731. [Google Scholar] [CrossRef] [PubMed]
  78. Le Moëne, O.; Ågmo, A. The neuroendocrinology of sexual attraction. Front. Neuroendocrinol. 2018, 51, 46–67. [Google Scholar] [CrossRef] [PubMed]
  79. Rodriguez, I.; Boehm, U. Pheromone Sensing in Mice. In Chemosensory Systems in Mammals, Fishes, and Insects; Springer: Berlin/Heidelberg, Germany, 2009; pp. 139–185. [Google Scholar] [CrossRef]
  80. Restrepo, D.; Arellano, J.; Oliva, A.M.; Schaefer, M.L.; Lin, W. Emerging views on the distinct but related roles of the main and accessory olfactory systems in responsiveness to chemosensory signals in mice. Horm. Behav. 2004, 46, 247–256. [Google Scholar] [CrossRef]
  81. Lin, D.Y.; Zhang, S.Z.; Block, E.; Katz, L.C. Encoding social signals in the mouse main olfactory bulb. Nature 2005, 434, 470–477. [Google Scholar] [CrossRef]
  82. Cotton, S. Individual Recognition: Mice, MUPs and the MHC. Curr. Biol. 2007, 17, R971–R973. [Google Scholar] [CrossRef]
  83. Overath, P.; Sturm, T.; Rammensee, H.-G. Of volatiles and peptides: In search for MHC-dependent olfactory signals in social communication. Cell. Mol. Life Sci. CMLS 2014, 71, 2429–2442. [Google Scholar] [CrossRef]
  84. Ruff, J.S.; Nelson, A.C.; Kubinak, J.L.; Potts, W.K. MHC signaling during social communication. Adv. Exp. Med. Biol. 2012, 738, 290–313. [Google Scholar]
  85. Ditchkoff, S.S.; Lochmiller, R.L.; Masters, R.E.; Hoofer, S.R.; Bussche, R.A.V.D. Major-histocompatibility-complex-associated variation in secondary sexual traits of white-tailed deer (Odocoileus virginianus): Evidence for good-genes advertisement. Evolution 2001, 55, 616–625. [Google Scholar] [CrossRef]
  86. Hurst, J.L.; Beynon, R.J. Rodent Urinary Proteins: Genetic Identity Signals and Pheromones. In Chemical Signals in Vertebrates 12; Springer: New York, NY, USA, 2012; pp. 117–133. [Google Scholar] [CrossRef]
  87. Weiss, J.; Vacher, H.; Trouillet, A.-C.; Leinders-Zufall, T.; Zufall, F.; Chamero, P. Sensing and avoiding sick conspecifics requires Gαi2 + vomeronasal neurons. BMC Biol. 2023, 21, 152. [Google Scholar] [CrossRef] [PubMed]
  88. Kavaliers, M.; Bishnoi, I.R.; Ossenkopp, K.-P.; Choleris, E. Progesterone and disgust: A response to “progesterone does raise disgust”. Horm. Behav. 2022, 137, 104936. [Google Scholar] [CrossRef] [PubMed]
  89. Millet, P.; Martin, T.; Opiekun, M.; Beauchamp, G.K.; Kimball, B.A. Differing Alterations of Odor Volatiles Among Pathogenic Stimuli. Chem. Senses 2021, 46, bjab030. [Google Scholar] [CrossRef]
  90. Kavaliers, M.; Colwell, D.D.; Ossenkopp, K.-P.; Perrot-Sinal, T.S. Altered Responses to Female Odors in Parasitized Male Mice: Neuromodulatory Mechanisms and Relations to Female Choice. Behav. Ecol. Sociobiol. 1997, 40, 373–384. [Google Scholar] [CrossRef]
  91. Gogolla, N. The insular cortex. Curr. Biol. 2017, 27, R580–R586. [Google Scholar] [CrossRef] [PubMed]
  92. Rogers-Carter, M.M.; Christianson, J.P. An insular view of the social decision-making network. Neurosci. Biobehav. Rev. 2019, 103, 119–132. [Google Scholar] [CrossRef] [PubMed]
  93. Soutschek, A. Neural Circuits Regulating Social Behavior: Highlighting the Causal Contribution of the Lateral Habenula. Biol. Psychiatry 2018, 83, 546–547. [Google Scholar] [CrossRef]
  94. Kang, N.; McCarthy, E.; Cherry, J.; Baum, M. A sex comparison of the anatomy and function of the main olfactory bulb–medial amygdala projection in mice. Neuroscience 2011, 172, 196–204. [Google Scholar] [CrossRef]
  95. Curtis, J.T.; Wang, Z. Forebrain c- fos expression under conditions conducive to pair bonding in female prairie voles (Microtus ochrogaster). Physiol. Behav. 2003, 80, 95–101. [Google Scholar] [CrossRef]
  96. Brennan, P.A.; Keverne, E.B. Something in the Air? New Insights into Mammalian Pheromones. Curr. Biol. 2004, 14, R81–R89. [Google Scholar] [CrossRef]
  97. Bergan, J.F.; Ben-Shaul, Y.; Dulac, C. Sex-specific processing of social cues in the medial amygdala. eLife 2014, 3, e02743. [Google Scholar] [CrossRef] [PubMed]
  98. Ben-Shaul, Y.; Katz, L.; Mooney, R.; Dulac, C. In vivo vomeronasal stimulation reveals sensory encoding of conspecific and allospecific cues by the mouse accessory olfactory bulb. Proc. Natl. Acad. Sci. USA 2010, 107, 5172–5177. [Google Scholar] [CrossRef] [PubMed]
  99. Raam, T.; Hong, W. Organization of neural circuits underlying social behavior: A consideration of the medial amygdala. Curr. Opin. Neurobiol. 2021, 68, 124–136. [Google Scholar] [CrossRef] [PubMed]
  100. Choi, G.B.; Dong, H.; Murphy, A.J.; Valenzuela, D.M.; Yancopoulos, G.D.; Swanson, L.W.; Anderson, D.J. Lhx6 Delineates a Pathway Mediating Innate Reproductive Behaviors from the Amygdala to the Hypothalamus. Neuron 2005, 46, 647–660. [Google Scholar] [CrossRef]
  101. Miller, S.M.; Marcotulli, D.; Shen, A.; Zweifel, L.S. Divergent medial amygdala projections regulate approach-avoidance conflict behavior. Nat. Neurosci. 2019, 22, 565–575. [Google Scholar] [CrossRef]
  102. Martinez, R.; Carvalho-Netto, E.; Ribeiro-Barbosa, É.; Baldo, M.V.; Canteras, N. Amygdalar roles during exposure to a live predator and to a predator-associated context. Neuroscience 2011, 172, 314–328. [Google Scholar] [CrossRef]
  103. Dielenberg, R.; Hunt, G.; McGregor, I. “When a rat smells a cat”: The distribution of Fos immunoreactivity in rat brain following exposure to a predatory odor. Neuroscience 2001, 104, 1085–1097. [Google Scholar] [CrossRef]
  104. Arakawa, H.; Arakawa, K.; Deak, T. Oxytocin and vasopressin in the medial amygdala differentially modulate approach and avoidance behavior toward illness-related social odor. Neuroscience 2010, 171, 1141–1151. [Google Scholar] [CrossRef]
  105. Kwon, J.-T.; Ryu, C.; Lee, H.; Sheffield, A.; Fan, J.; Cho, D.H.; Bigler, S.; Sullivan, H.A.; Choe, H.K.; Wickersham, I.R.; et al. An amygdala circuit that suppresses social engagement. Nature 2021, 593, 114–118. [Google Scholar] [CrossRef]
  106. Rogers-Carter, M.M.; Varela, J.A.; Gribbons, K.B.; Pierce, A.F.; McGoey, M.T.; Ritchey, M.; Christianson, J.P. Insular cortex mediates approach and avoidance responses to social affective stimuli. Nat. Neurosci. 2018, 21, 404–414. [Google Scholar] [CrossRef]
  107. Butti, C.; Hof, P.R. The insular cortex: A comparative perspective. Brain Struct. Funct. 2010, 214, 477–493. [Google Scholar] [CrossRef] [PubMed]
  108. Gehrlach, D.A.; Weiand, C.; Gaitanos, T.N.; Cho, E.; Klein, A.S.; Hennrich, A.A.; Conzelmann, K.-K.; Gogolla, N. A whole-brain connectivity map of mouse insular cortex. eLife 2020, 9, e55585. [Google Scholar] [CrossRef] [PubMed]
  109. Min, J.-Y.; Park, S.; Cho, J.; Huh, Y. The anterior insular cortex processes social recognition memory. Sci. Rep. 2023, 13, 10853. [Google Scholar] [CrossRef] [PubMed]
  110. Rieger, N.S.; Worley, N.B.; Ng, A.J.; Christianson, J.P. Insular cortex modulates social avoidance of sick rats. Behav. Brain Res. 2022, 416, 113541. [Google Scholar] [CrossRef] [PubMed]
  111. Dolensek, N.; Gehrlach, D.A.; Klein, A.S.; Gogolla, N. Facial expressions of emotion states and their neuronal correlates in mice. Science 2020, 368, 89–94. [Google Scholar] [CrossRef] [PubMed]
  112. Pardo-Bellver, C.; Cádiz-Moretti, B.; Novejarque, A.; Martínez-García, F.; Lanuza, E. Differential efferent projections of the anterior, posteroventral, and posterodorsal subdivisions of the medial amygdala in mice. Front. Neuroanat. 2012, 6, 33. [Google Scholar] [CrossRef] [PubMed]
  113. Kargl, D.; Kaczanowska, J.; Ulonska, S.; Groessl, F.; Piszczek, L.; Lazovic, J.; Buehler, K.; Haubensak, W. The amygdala instructs insular feedback for affective learning. eLife 2020, 9, e60336. [Google Scholar] [CrossRef]
  114. Kayyal, H.; Yiannakas, A.; Kolatt Chandran, S.; Khamaisy, M.; Sharma, V.; Rosenblum, K. Activity of Insula to Basolateral Amygdala Projecting Neurons is Necessary and Sufficient for Taste Valence Representation. J. Neurosci. 2019, 39, 9369–9382. [Google Scholar] [CrossRef]
  115. Namboodiri, V.M.K.; Rodriguez-Romaguera, J.; Stuber, G.D. The habenula. Curr. Biol. 2016, 26, R873–R877. [Google Scholar] [CrossRef]
  116. Flanigan, M.; Aleyasin, H.; Takahashi, A.; Golden, S.A.; Russo, S.J. An emerging role for the lateral habenula in aggressive behavior. Pharmacol. Biochem. Behav. 2017, 162, 79–86. [Google Scholar] [CrossRef]
  117. Wang, D.; Li, Y.; Feng, Q.; Guo, Q.; Zhou, J.; Luo, M. Learning shapes the aversion and reward responses of lateral habenula neurons. eLife 2017, 6, e23045. [Google Scholar] [CrossRef] [PubMed]
  118. Benekareddy, M.; Stachniak, T.J.; Bruns, A.; Knoflach, F.; von Kienlin, M.; Künnecke, B.; Ghosh, A. Identification of a Corticohabenular Circuit Regulating Socially Directed Behavior. Biol. Psychiatry 2018, 83, 607–617. [Google Scholar] [CrossRef] [PubMed]
  119. He, Z.-X.; Xi, K.; Liu, K.-J.; Yue, M.-H.; Wang, Y.; Yin, Y.-Y.; Liu, L.; He, X.-X.; Yu, H.-L.; Xing, Z.-K.; et al. A Nucleus Accumbens Tac1 Neural Circuit Regulates Avoidance Responses to Aversive Stimuli. Int. J. Mol. Sci. 2023, 24, 4346. [Google Scholar] [CrossRef] [PubMed]
  120. Klawonn, A.M.; Malenka, R.C. Nucleus accumbens modulation in reward and aversion. In Cold Spring Harbor Symposia on Quantitative Biology; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 2018; Volume 83, pp. 119–129. [Google Scholar]
  121. Britt, J.P.; Benaliouad, F.; McDevitt, R.A.; Stuber, G.D.; Wise, R.A.; Bonci, A. Synaptic and Behavioral Profile of Multiple Glutamatergic Inputs to the Nucleus Accumbens. Neuron 2012, 76, 790–803. [Google Scholar] [CrossRef]
  122. Russo, S.J.; Nestler, E.J. The brain reward circuitry in mood disorders. Nat. Rev. Neurosci. 2013, 14, 609–625. [Google Scholar] [CrossRef]
  123. de Jong, J.W.; Afjei, S.A.; Dorocic, I.P.; Peck, J.R.; Liu, C.; Kim, C.K.; Tian, L.; Deisseroth, K.; Lammel, S. A neural circuit mechanism for encoding aversive stimuli in the mesolimbic dopamine system. Neuron 2019, 101, 133–151. [Google Scholar] [CrossRef]
  124. Zhou, K.; Xu, H.; Lu, S.; Jiang, S.; Hou, G.; Deng, X.; He, M.; Zhu, Y. Reward and aversion processing by input-defined parallel nucleus accumbens circuits in mice. Nat. Commun. 2022, 13, 6244. [Google Scholar] [CrossRef]
  125. Dölen, G.; Darvishzadeh, A.; Huang, K.W.; Malenka, R.C. Social reward requires coordinated activity of nucleus accumbens oxytocin and serotonin. Nature 2013, 501, 179–184. [Google Scholar] [CrossRef]
  126. Damsma, G.; Pfaus, J.G.; Wenkstern, D.; Phillips, A.G.; Fibiger, H.C. Sexual behavior increases dopamine transmission in the nucleus accumbens and striatum of male rats: Comparison with novelty and locomotion. Behav. Neurosci. 1992, 106, 181. [Google Scholar] [CrossRef]
  127. Guadarrama-Bazante, I.L.; Rodríguez-Manzo, G. Nucleus accumbens dopamine increases sexual motivation in sexually satiated male rats. Psychopharmacology 2019, 236, 1303–1312. [Google Scholar] [CrossRef]
  128. Dai, B.; Sun, F.; Tong, X.; Ding, Y.; Kuang, A.; Osakada, T.; Li, Y.; Lin, D. Responses and functions of dopamine in nucleus accumbens core during social behaviors. Cell Rep. 2022, 40, 111246. [Google Scholar] [CrossRef] [PubMed]
  129. Cheng, J.Y.; Zhang, S.W.; Tai, F.D. Effects of nucleus accumbens oxytocin and its antagonist on social approach behavior. Behav. Pharmacol. 2016, 27, 672–680. [Google Scholar]
  130. Dölen, G.; Malenka, R.C. The emerging role of nucleus accumbens oxytocin in social cognition. Biol. Psychiatry 2014, 76, 354–355. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, Y.; Wang, Z.X. Nucleus accumbens oxytocin and dopamine interact to regulate pair bond formation in female prairie voles. Neuroscience 2003, 121, 537–544. [Google Scholar] [CrossRef] [PubMed]
  132. Quintana, G.R.; Mac Cionnaith, C.E.; Pfaus, J.G. Behavioral, neural, and molecular mechanisms of conditioned mate preference: The role of opioids and first experiences of sexual reward. Int. J. Mol. Sci. 2022, 23, 8928. [Google Scholar] [CrossRef]
  133. Pitchers, K.K.; Frohmader, K.S.; Vialou, V.; Mouzon, E.; Nestler, E.J.; Lehman, M.N.; Coolen, L.M. ΔFosB in the nucleus accumbens is critical for reinforcing effects of sexual reward. Genes Brain Behav. 2010, 9, 831–840. [Google Scholar] [CrossRef] [PubMed]
  134. Tseng, Y.T.; Schaefke, B.; Wei, P.; Wang, L. Defensive responses: Behaviour, the brain and the body. Nat. Rev. Neurosci. 2023, 24, 655–671. [Google Scholar] [CrossRef]
  135. Martin-Fernandez, M.; Menegolla, A.P.; Lopez-Fernandez, G.; Winke, N.; Jercog, D.; Kim, H.-R.; Girard, D.; Dejean, C.; Herry, C. Prefrontal circuits encode both general danger and specific threat representations. Nat. Neurosci. 2023, 26, 2147–2157. [Google Scholar] [CrossRef]
  136. Leschak, C.J.; Hornstein, E.A.; Haltom, K.E.B.; Johnson, K.L.; Breen, E.C.; Irwin, M.R.; Eisenberger, N.I. Ventromedial prefrontal cortex activity differentiates sick from healthy faces: Associations with inflammatory responses and disease avoidance motivation. Brain Behav. Immun. 2022, 100, 48–54. [Google Scholar] [CrossRef]
  137. Keller, J.K.; Wülfing, C.; Wahl, J.; Diekhof, E.K. Disease-related disgust promotes antibody release in human saliva. Brain Behav. Immun. Health 2022, 24, 100489. [Google Scholar] [CrossRef]
  138. Schaller, M.; Miller, G.E.; Gervais, W.M.; Yager, S.; Chen, E. Mere Visual Perception of Other People’s Disease Symptoms Facilitates a More Aggressive Immune Response. Psychol. Sci. 2010, 21, 649–652. [Google Scholar] [CrossRef] [PubMed]
  139. Truby, N.L.; Kim, R.K.; Silva, G.M.; Qu, X.; Picone, J.A.; Alemu, R.; Neve, R.L.; Cui, X.; Liu, J.; Hamilton, P.J. A zinc finger transcription factor tunes social behaviors by controlling transposable elements and immune response in prefrontal cortex. bioRxiv 2023. [Google Scholar] [CrossRef]
  140. Filiano, A.J.; Xu, Y.; Tustison, N.J.; Marsh, R.L.; Baker, W.; Smirnov, I.; Overall, C.C.; Gadani, S.P.; Turner, S.D.; Weng, Z.; et al. Unexpected role of interferon-γ in regulating neuronal connectivity and social behaviour. Nature 2016, 535, 425–429. [Google Scholar] [CrossRef]
  141. Archie, E.A.; Tung, J. Social behavior and the microbiome. Curr. Opin. Behav. Sci. 2015, 6, 28–34. [Google Scholar] [CrossRef]
  142. Bleich, A.; Hansen, A.K. Time to include the gut microbiota in the hygienic standardisation of laboratory rodents. Comp. Immunol. Microbiol. Infect. Dis. 2012, 35, 81–92. [Google Scholar] [CrossRef] [PubMed]
  143. Hansen, A.K.; Krych, Ł.; Nielsen, D.S.; Hansen, C.H.F. A Review of Applied Aspects of Dealing with Gut Microbiota Impact on Rodent Models. ILAR J. 2015, 56, 250–264. [Google Scholar] [CrossRef]
  144. Belkaid, Y.; Segre, J.A. Dialogue between skin microbiota and immunity. Science 2014, 346, 954–959. [Google Scholar] [CrossRef]
  145. Sherwin, E.; Bordenstein, S.R.; Quinn, J.L.; Dinan, T.G.; Cryan, J.F. Microbiota and the social brain. Science 2019, 366, 587. [Google Scholar] [CrossRef]
  146. Lavrinienko, A.; Tukalenko, E.; Mappes, T.; Watts, P.C. Skin and gut microbiomes of a wild mammal respond to different environmental cues. Microbiome 2018, 6, 209. [Google Scholar] [CrossRef]
  147. Archie, E.A.; Theis, K.R. Animal behaviour meets microbial ecology. Anim. Behav. 2011, 82, 425–436. [Google Scholar] [CrossRef]
  148. Ezenwa, V.O.; Williams, A.E. Microbes and animal olfactory communication: Where do we go from here? BioEssays 2014, 36, 847–854. [Google Scholar] [CrossRef]
  149. Sharon, G.; Segal, D.; Ringo, J.M.; Hefetz, A.; Zilber-Rosenberg, I.; Rosenberg, E.; Collier, R.J. Commensal bacteria play a role in mating preference of Drosophila melanogaster. Proc. Natl. Acad. Sci. USA 2010, 107, 20051–20056. [Google Scholar] [CrossRef] [PubMed]
  150. Li, Q.; Korzan, W.J.; Ferrero, D.M.; Chang, R.B.; Roy, D.S.; Buchi, M.; Lemon, J.K.; Kaur, A.W.; Stowers, L.; Fendt, M.; et al. Synchronous Evolution of an Odor Biosynthesis Pathway and Behavioral Response. Curr. Biol. 2013, 23, 11–20. [Google Scholar] [CrossRef] [PubMed]
  151. Yi, X.; Cha, M. Gut Dysbiosis Has the Potential to Reduce the Sexual Attractiveness of Mouse Female. Front. Microbiol. 2022, 13, 916766. [Google Scholar] [CrossRef] [PubMed]
  152. Ueyama, J.; Nadai, M.; Kanazawa, H.; Iwase, M.; Nakayama, H.; Hashimoto, K.; Yokoi, T.; Baba, K.; Takagi, K.; Takagi, K.; et al. Endotoxin from various gram-negative bacteria has differential effects on function of hepatic cytochrome P450 and drug transporters. Eur. J. Pharmacol. 2005, 510, 127–134. [Google Scholar] [CrossRef]
  153. Baker, J.M.; Al-Nakkash, L.; Herbst-Kralovetz, M.M. Estrogen–gut microbiome axis: Physiological and clinical implications. Maturitas 2017, 103, 45–53. [Google Scholar] [CrossRef] [PubMed]
  154. Kaliannan, K.; Robertson, R.C.; Murphy, K.; Stanton, C.; Kang, C.; Wang, B.; Hao, L.; Bhan, A.K.; Kang, J.X. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome 2018, 6, 205. [Google Scholar] [CrossRef]
  155. Org, E.; Mehrabian, M.; Parks, B.W.; Shipkova, P.; Liu, X.; Drake, T.A.; Lusis, A.J. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes 2016, 7, 313–322. [Google Scholar] [CrossRef]
  156. Yurkovetskiy, L.; Burrows, M.; Khan, A.A.; Graham, L.; Volchkov, P.; Becker, L.; Antonopoulos, D.; Umesaki, Y.; Chervonsky, A.V. Gender Bias in Autoimmunity Is Influenced by Microbiota. Immunity 2013, 39, 400–412. [Google Scholar] [CrossRef]
  157. Markle, J.G.M.; Frank, D.N.; Mortin-Toth, S.; Robertson, C.E.; Feazel, L.M.; Rolle-Kampczyk, U.; von Bergen, M.; McCoy, K.D.; Macpherson, A.J.; Danska, J.S. Sex Differences in the Gut Microbiome Drive Hormone-Dependent Regulation of Autoimmunity. Science 2013, 339, 1084–1088. [Google Scholar] [CrossRef]
  158. Zeibich, L.; Koebele, S.V.; Bernaud, V.E.; Ilhan, Z.E.; Dirks, B.; Northup-Smith, S.N.; Neeley, R.; Maldonado, J.; Nirmalkar, K.; Files, J.A.; et al. Surgical Menopause and Estrogen Therapy Modulate the Gut Microbiota, Obesity Markers, and Spatial Memory in Rats. Front. Cell. Infect. Microbiol. 2021, 11, 702628. [Google Scholar] [CrossRef]
  159. Acharya, K.D.; Noh, H.L.; Graham, M.E.; Suk, S.; Friedline, R.H.; Gomez, C.C.; Parakoyi, A.E.R.; Chen, J.; Kim, J.K.; Tetel, M.J. Distinct Changes in Gut Microbiota Are Associated with Estradiol-Mediated Protection from Diet-Induced Obesity in Female Mice. Metabolites 2021, 11, 499. [Google Scholar] [CrossRef]
  160. de Catanzaro, D. Sex steroids as pheromones in mammals: The exceptional role of estradiol. Horm. Behav. 2015, 68, 103–116. [Google Scholar] [CrossRef] [PubMed]
  161. Pietras, R.J. Sex pheromone production by preputial gland: The regulatory role of estrogen. Chem. Senses 1981, 6, 391–408. [Google Scholar] [CrossRef]
  162. Bouilloud, M.; Galan, M.; Dubois, A.; Diagne, C.; Marianneau, P.; Roche, B.; Charbonnel, N. Three-way relationships between gut microbiota, helminth assemblages and bacterial infections in wild rodent populations. Peer Community J. 2023, 3, e18. [Google Scholar] [CrossRef]
  163. Zhao, Y.; Yang, S.; Li, B.; Li, W.; Wang, J.; Chen, Z.; Yang, J.; Tan, H.; Li, J. Alterations of the Mice Gut Microbiome via Schistosoma japonicum Ova-Induced Granuloma. Front. Microbiol. 2019, 10, 352. [Google Scholar] [CrossRef] [PubMed]
  164. Pandian, J. Analysis of Wild Rodent Gut Microbiota as a Function of Exposure to Ticks and Tick-borne Pathogens. Unbuplished Honor. Theses 2023, 1683. Available online: https://scholarship.richmond.edu/honors-theses/1683 (accessed on 3 January 2024).
  165. Aivelo, T.; Norberg, A. Parasite–microbiota interactions potentially affect intestinal communities in wild mammals. J. Anim. Ecol. 2018, 87, 438–447. [Google Scholar] [CrossRef]
  166. Reynolds, L.A.; Smith, K.A.; Filbey, K.J.; Harcus, Y.; Hewitson, J.P.; Redpath, S.A.; Valdez, Y.; Yebra, M.J.; Finlay, B.B.; Maizels, R.M. Commensal-pathogen interactions in the intestinal tract: Lactobacilli promote infection with, and are promoted by, helminth parasites. Gut Microbes 2014, 5, 522–532. [Google Scholar] [CrossRef]
  167. Su, C.; Su, L.; Li, Y.; Long, S.R.; Chang, J.; Zhang, W.; Walker, W.A.; Xavier, R.J.; Cherayil, B.J.; Shi, H.N. Helminth-induced alterations of the gut microbiota exacerbate bacterial colitis. Mucosal Immunol. 2018, 11, 144–157. [Google Scholar] [CrossRef]
  168. Delbare, S.Y.N.; Ahmed-Braimah, Y.H.; Wolfner, M.F.; Clark, A.G. Interactions between the microbiome and mating influence the female’s transcriptional profile in Drosophila melanogaster. Sci. Rep. 2020, 10, 18168. [Google Scholar] [CrossRef]
  169. Arbuthnott, D.; Levin, T.C.; Promislow, D.E.L. Impacts of Wolbachia and the microbiome on mate choice in Drosophila melanogaster. J. Evol. Biol. 2016, 29, 461–468. [Google Scholar] [CrossRef] [PubMed]
  170. Winslow, J.T.; Ferguson, J.N.; Young, L.J.; Hearn, E.F.; Matzuk, M.M.; Insel, T.R. Social amnesia in mice lacking the oxytocin gene. Nat. Genet. 2000, 25, 284–288. [Google Scholar] [CrossRef]
  171. Young, L.J. The neurobiology of social recognition, approach, and avoidance. Biol. Psychiatry 2002, 51, 18–26. [Google Scholar] [CrossRef] [PubMed]
  172. Kavaliers, M.; Ossenkopp, K.; Choleris, E. Social neuroscience of disgust. Genes Brain Behav. 2019, 18, e12508. [Google Scholar] [CrossRef] [PubMed]
  173. Goodson, J.L. Deconstructing sociality, social evolution and relevant nonapeptide functions. Psychoneuroendocrinology 2013, 38, 465–478. [Google Scholar] [CrossRef] [PubMed]
  174. Marlin, B.J.; Froemke, R.C. Oxytocin modulation of neural circuits for social behavior. Dev. Neurobiol. 2017, 77, 169–189. [Google Scholar] [CrossRef]
  175. Kavaliers, M.; Choleris, E.; Ågmo, A.; Pfaff, D.W. Olfactory-mediated parasite recognition and avoidance: Linking genes to behavior. Horm. Behav. 2004, 46, 272–283. [Google Scholar] [CrossRef]
  176. Boulet, N.P.; Cloutier, C.J.; Ossenkopp, K.-P.; Kavaliers, M. Oxytocin, social factors, and the expression of conditioned disgust (Anticipatory nausea) in male rats. Behav. Pharmacol. 2016, 27, 718–725. [Google Scholar] [CrossRef]
  177. Paletta, P.; Bass, N.; Kavaliers, M.; Choleris, E. The role of oxytocin in shaping complex social behaviours: Possible interactions with other neuromodulators. Philos. Trans. R. Soc. B 2022, 377, 20210058. [Google Scholar] [CrossRef]
  178. Yoest, K.E.; Cummings, J.A.; Becker, J.B. Ovarian Hormones Mediate Changes in Adaptive Choice and Motivation in Female Rats. Front. Behav. Neurosci. 2019, 13, 250. [Google Scholar] [CrossRef]
  179. Thompson, T.L.; Moss, R.L. Modulation of mesolimbic dopaminergic activity over the rat estrous cycle. Neurosci. Lett. 1997, 229, 145–148. [Google Scholar] [CrossRef] [PubMed]
  180. Choleris, E.; Clipperton-Allen, A.E.; Phan, A.; Valsecchi, P.; Kavaliers, M. Estrogenic involvement in social learning, social recognition and pathogen avoidance. Front. Neuroendocrinol. 2012, 33, 140–159. [Google Scholar] [CrossRef] [PubMed]
  181. Kavaliers, M.; Choleris, E.; Tenk, C.M.; Pfaff, D.W.; Ogawa, S. Estrogen receptor α and β involvement in the mediation of the aversive responses of female mice to the volatile and involatile odors of parasitized males. Soc. Behav. Neuroendocrinol. Abstr. 2008, 33, 634–642. [Google Scholar]
  182. Lymer, J.M.; Sheppard, P.A.S.; Kuun, T.; Blackman, A.; Jani, N.; Mahbub, S.; Choleris, E. Estrogens and their receptors in the medial amygdala rapidly facilitate social recognition in female mice. Psychoneuroendocrinology 2018, 89, 30–38. [Google Scholar] [CrossRef]
  183. Sexton, C. Effects of an Oxytocin Receptor Antagonist on Estrogens’ Facilitative Effects on Social Recognition in the Medial Amygdala on Female Mice. Master’s Thesis, Department of Psychology, University of Guelph, Guelph, ON, Canada, 2022. [Google Scholar]
  184. Arakawa, H.; Cruz, S.; Deak, T. Attractiveness of illness-associated odorant cues in female rats is modulated by ovarian hormones, but not associated with pro-inflammatory cytokine levels. Brain Behav. Immun. 2012, 26, 40–49. [Google Scholar] [CrossRef]
  185. Brown, C.M.; Mulcahey, T.A.; Filipek, N.C.; Wise, P.M. Production of Proinflammatory Cytokines and Chemokines During Neuroinflammation: Novel Roles for Estrogen Receptors α and β. Endocrinology 2010, 151, 4916–4925. [Google Scholar] [CrossRef]
  186. Seredynski, A.L.; Balthazart, J.; Christophe, V.J.; Ball, G.F.; Cornil, C.A. Neuroestrogens rapidly regulate sexual motivation but not performance. J. Neurosci. 2013, 33, 164–174. [Google Scholar] [CrossRef]
  187. Ogawa, S.; Eng, V.; Taylor, J.; Lubahn, D.B.; Korach, K.S.; Pfaff, D.W. Roles of estrogen receptor-alpha gene expression in reproduction-related behaviors in female mice. Endocrinology 1998, 139, 5070–5081. [Google Scholar] [CrossRef]
  188. Fleischman, D.S.; Fessler, D.M.T. Progesterone’s effects on the psychology of disease avoidance: Support for the compensatory behavioral prophylaxis hypothesis. Horm. Behav. 2011, 59, 271–275. [Google Scholar] [CrossRef]
  189. Kavaliers, M.; Bishnoi, I.R.; Ossenkopp, K.-P.; Choleris, E. Differential effects of progesterone on social recognition and the avoidance of pathogen threat by female mice. Horm. Behav. 2021, 127, 104873. [Google Scholar] [CrossRef]
  190. Bressan, P.; Kramer, P. Progesterone does raise disgust. Horm. Behav. 2022, 137, 104937. [Google Scholar] [CrossRef] [PubMed]
  191. Lopes, P.C. Anticipating infection: How parasitism risk changes animal physiology. Funct. Ecol. 2023, 37, 821–830. [Google Scholar] [CrossRef]
  192. Curtis, V.A. Infection-avoidance behaviour in humans and other animals. Trends Immunol. 2014, 35, 457–464. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Diagram illustrating the sectional progression of the review. Section (i) discusses aspects of mate choice and the avoidance of pathogen threat processed in conspecific cues. Section (ii) discusses the detection of the pathogen risk and olfaction. Section (iii) discusses the neurobiological underpinnings associated with appropriate affective and cognitive responses to pathogenic cues, with a particular consideration of brain regions, hormones, and neuropeptide systems. Created with BioRender.com.
Figure 1. Diagram illustrating the sectional progression of the review. Section (i) discusses aspects of mate choice and the avoidance of pathogen threat processed in conspecific cues. Section (ii) discusses the detection of the pathogen risk and olfaction. Section (iii) discusses the neurobiological underpinnings associated with appropriate affective and cognitive responses to pathogenic cues, with a particular consideration of brain regions, hormones, and neuropeptide systems. Created with BioRender.com.
Animals 14 00296 g001
Figure 2. Regions and projections in the mouse brain that are proposed to be involved in the processing and mediation of the behavioural response to the pathogen risk. It is likely that additional systems involved in social information processing are also involved. MOE (main olfactory epithelium); VNO (vomeronasal organ); AOB (accessory olfactory bulb); mPFC (medial prefrontal cortex); NAc (nucleus accumbens); Hb (habenula); Amy (amygdala).
Figure 2. Regions and projections in the mouse brain that are proposed to be involved in the processing and mediation of the behavioural response to the pathogen risk. It is likely that additional systems involved in social information processing are also involved. MOE (main olfactory epithelium); VNO (vomeronasal organ); AOB (accessory olfactory bulb); mPFC (medial prefrontal cortex); NAc (nucleus accumbens); Hb (habenula); Amy (amygdala).
Animals 14 00296 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cantini, D.; Choleris, E.; Kavaliers, M. Neurobiology of Pathogen Avoidance and Mate Choice: Current and Future Directions. Animals 2024, 14, 296. https://doi.org/10.3390/ani14020296

AMA Style

Cantini D, Choleris E, Kavaliers M. Neurobiology of Pathogen Avoidance and Mate Choice: Current and Future Directions. Animals. 2024; 14(2):296. https://doi.org/10.3390/ani14020296

Chicago/Turabian Style

Cantini, Dante, Elena Choleris, and Martin Kavaliers. 2024. "Neurobiology of Pathogen Avoidance and Mate Choice: Current and Future Directions" Animals 14, no. 2: 296. https://doi.org/10.3390/ani14020296

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