Next Article in Journal
Bacterial Species Involved in Venous Leg Ulcer Infections and Their Sensitivity to Antibiotherapy—An Alarm Signal Regarding the Seriousness of Chronic Venous Insufficiency C6 Stage and Its Need for Prompt Treatment
Next Article in Special Issue
Developing the Common Marmoset as a Translational Geroscience Model to Study the Microbiome and Healthy Aging
Previous Article in Journal
Using Fungi in Artificial Microbial Consortia to Solve Bioremediation Problems
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Early-Life Cecal Microbiota Transplantation on Social Stress and Injurious Behaviors in Egg-Laying Chickens

by
Yuechi Fu
1,
Jiaying Hu
1,
Huanmin Zhang
2,
Marisa A. Erasmus
1,
Timothy A. Johnson
1 and
Heng-Wei Cheng
3,*
1
Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
2
U.S. National Poultry Research Center, USDA-ARS, Athens, GA 30605, USA
3
Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(3), 471; https://doi.org/10.3390/microorganisms12030471
Submission received: 26 January 2024 / Revised: 21 February 2024 / Accepted: 23 February 2024 / Published: 26 February 2024
(This article belongs to the Special Issue Fecal Microbiota Transplantation in Animals)

Abstract

:

Simple Summary

A plethora of studies have evidenced that the gut microbiota profoundly influences host brain function and behavioral characteristics in humans and various animals. In laying hens, it has been reported that injurious behaviors (such as aggressive pecking, feather pecking, and cannibalism) are associated with dysregulation of the microbiota–gut–brain axis. This study further investigated the effects of the early-life transplantation of different cecal contents on aggressiveness and related behaviors in chickens. Cecal bacterial profiles of two divergently selected inbred genetic lines (donors) were analyzed and then orally transferred separately into newly hatched male chicks of a commercial layer strain (recipients). Effects of cecal microbiota transplantation on growth, physiology, and behavior were examined in the recipient chicks. This study first evidenced that social stress and stress-related injurious behaviors in chickens can be reduced by modification of the gut microbiota composition and brain serotonergic activities via the gut–brain axis. The results provide new insights into understanding the cellular mechanisms of the gut microbiota in regulating stress-induced abnormal behaviors and offer a novel strategy for improving health and welfare in laying hens.

Abstract

Injurious behaviors (i.e., aggressive pecking, feather pecking, and cannibalism) in laying hens are a critical issue facing the egg industry due to increased social stress and related health and welfare issues as well as economic losses. In humans, stress-induced dysbiosis increases gut permeability, releasing various neuroactive factors, causing neuroinflammation and related neuropsychiatric disorders via the microbiota–gut–brain axis, and consequently increasing the frequency and intensity of aggression and violent behaviors. Restoration of the imbalanced gut microbial composition has become a novel treatment strategy for mental illnesses, such as depression, anxiety, bipolar disorder, schizophrenia, impulsivity, and compulsivity. A similar function of modulating gut microbial composition following stress challenge may be present in egg-laying chickens. The avian cecum, as a multi-purpose organ, has the greatest bacterial biodiversity (bacterial diversity, richness, and species composition) along the gastrointestinal tract, with vitally important functions in maintaining physiological and behavioral homeostasis, especially during the periods of stress. To identify the effects of the gut microbiome on injurious behaviors in egg-laying chickens, we have designed and tested the effects of transferring cecal contents from two divergently selected inbred chicken lines on social stress and stress-related injurious behaviors in recipient chicks of a commercial layer strain. This article reports the outcomes from a multi-year study on the modification of gut microbiota composition to reduce injurious behaviors in egg-laying chickens. An important discovery of this corpus of experiments is that injurious behaviors in chickens can be reduced or inhibited through modifying the gut microbiota composition and brain serotonergic activities via the gut–brain axis, without donor-recipient genetic effects.

1. Introduction

Domestic egg-laying chickens have been continuously selected for high egg production with a high feed efficiency to meet the constant increase in human nutrition demand for eggs due to both population growth and rising individual consumption [1,2]. However, extreme selection is often at the expense of the animal’s health and welfare [3,4]; i.e., selecting one trait (such as production) could affect other traits, causing negative impacts on the animals [5]. Based on the traditional selection theory, an animal’s productivity is correlated with its competitive ability [6,7]. As unexpected results, the effects of selection for increased production reportedly resulted in increased interspecific competition and aggression [8,9,10]. In one of our previous studies, egg production increased significantly in former commercial Dekalb XL hens through more than 20 years of selection, while mortality associated with aggression and related injurious behaviors (aggressive pecking, severe feather pecking (SFP), and cannibalism) in non-beak trimmed hens also increased about 10-fold [11]. Increased injurious behaviors could be related to selection unequally affecting the animals’ adaptability to their rearing environments and management practices. Within a socioecological environment, not all animal individuals have an equal ability to modify their physiological and behavioral characteristics (such as personality traits for boldness, activity, and aggressiveness) in response to practice-associated stressors (inter-individual differences in adaption) [12,13,14,15]. Based on a dominance hierarchy or a ranking order, subordinates that are in direct contention with a dominant individual within a social group (i.e., the interactions between dominant higher-ranking (alpha) animals and submissive lower-ranking (beta) animals) exhibit fear, reducing their adaptation to the rearing environments and related management practices. Consequently, the subordinates enter a ‘pre-pathological state’ or ‘pathological state’ with physiological and metabolic disturbances [16,17,18]. Dominant chickens then exhibit an increased frequency of aggression and related injurious behaviors via the brain award systems and reinforced learning pathways, which could be similar to the brain systems reported in humans [19].
Aggression in chickens, as in most other species of social animals, is a highly complex social behavior. From an evolutionary viewpoint, aggression, as a natural part of an animal’s life, is essential for the animal to establish and maintain social status, to protect valuable resources (food and territory), and to reproduce successfully (survival, growth, breeding, and rearing offspring) [20,21,22,23]. However, some forms of aggression in chickens, such as excessive aggression-related injurious behaviors, can be harmful, leading to devastating consequences with increased social stress, feather and body damage, and injury (leading to cannibalism) [24,25,26]. In addition, numerous studies focusing on the function of gut microbiota in behavioral development have indicated that the dysregulation of the microbiota–gut–brain (MGB) axis has been implicated in abnormal behaviors (aggressive pecking, feather pecking (FP), and cannibalism) in laying hens [27,28]. Feather pecking may not be associated with dominance status; however, recent studies suggested that FP is related to social-stress-associated fearfulness [29]. Injurious behaviors, as a socially transmitted learning behavior, can be spread among flocks [30]. It has been previously reported that FP could affect up to 80% of birds in current housing environments [25].
Those injurious behaviors may be reduced through genetic selection [31,32,33,34]. However, there is “no sign that breeders will be able to guarantee the ‘non-peck’ layers in time” for hens to be housed in cage-free systems [35,36]. Egg production facilities are transferring from the conventional (battery) cage system to cage-free systems in the United States. Approximately 230 corporate customers, such as McDonald’s, Walmart, Subway, and Kroger, have pledged to only buy cage-free eggs by or before 2025. In addition, recent studies showed that selection for low-FP chickens failed to eliminate FP completely in flocks [37], which suggests that genetic selection should be paired with other management strategies [38]. Currently, beak trimming (BT), a routine procedure practiced in the United States egg industry, is the most effective method for reducing social stress by preventing and/or inhibiting injurious behaviors. However, BT has been criticized for causing tissue damage and pain (acute, chronic, or both) [39,40], negatively impacting the welfare of billions of chickens annually [41,42]. In addition, the chicken beak is a multipurpose organ playing a vital role in a variety of functions, from eating to defense against predators and parasites [43]. Beak trimming damages these beak functions, leading to abnormal behaviors and frustration [44]. Considerable concerns from the public have led to a growing global movement against the procedures causing pain and suffering in farm animals. However, recent studies have reported that FP and cannibalism still occur in beak-trimmed, floor-reared, and cage-free flocks [45]. Based on the outcomes, several studies have advocated that “solutions have to be found before thinking about banning BT” [45,46]. In addition, recent studies have indicated that FP and foraging are uncorrelated, which is inconsistent with the original hypothesis that FP is redirected food-related foraging pecks [47]. Feather pecking can lead to cannibalistic pecking, consequently eating and removing flesh from the victims by further reinforcing the behavior via the gut–brain reward systems (the central serotonergic and dopaminergic systems) [48,49]. In addition, injurious-behavior-associated social stress can disturb intestinal bacterial balance, resulting in physiological and behavioral disorders via the MGB axis [50,51].
The gut microbiota plays a critical role in early programming and later activity of the central stress systems, i.e., the hypothalamic–pituitary–adrenal (HPA) and the sympathetic–adrenal–medullary (SAM) axes [52,53,54]. Like an endocrine organ, the gut microbiota is sensitive and reactive to various exogenous stimuli, functioning as an environmental sensor linked to the pathogenesis of stress-related illnesses through the bidirectional communication of the MGB axis [55,56,57,58,59] and the microbiota–gut–immune (MGI) axis [60,61] in various animals including chickens [62,63,64]. Maintaining gut microbiota balance and health is essential for animals (including chickens) to maintain their optimal physiological and behavioral functions of growth, reproduction, and welfare. In humans, various psychological (emotional and mental overstimulation) and/or physical (environmental conditions) stressors alter gut microbiota diversity, composition, or both and increase the inability to maintain a healthy gut microbial profile, leading to neuropsychiatric disorders [65,66,67,68,69,70,71]. Targeting the intestinal microbiota with the goal of restoring its balance has been recognized as a novel therapeutic option for patients with neuropsychiatric disorders [72,73,74]. Several probiotics, as psychobiotics, such as Bifidobacterium and Lactobacillus, which can benefit mental health, have been used for preventing and treating patients with behavioral impairment, such as anxiety, depression, and impulsively and compulsively disrupted social behavior, via regulating the MGB, MGI, or both axes [75,76,77,78,79,80,81,82,83,84]. However, the use of purified probiotics benefits has shown mixed results, with several weaknesses including transient beneficial effects, requiring continuous administration over time due to the host’s resident microbial populations and “colonization resistance” [85]. Thus, it has been proposed that using live commensals coming directly from a healthy gut may be more effective than probiotics [86,87]. However, this hypothesis has not been well investigated in chickens.
The avian cecum, as a multipurpose organ, has a greater biological role than the cecum in most mammals [88,89,90,91]. In addition, chicken lines’ differences in the cecal microbiota composition in response to environmental stressors (such as ambient stress) [92] and experimental challenge models [93] have been reported. For example, laying hens showing high or low FP have different gut microbial populations [94,95] and intestinal and peripheral metabolite profiles [96,97]. However, a recent study reported that these differences may not be associated with FP and antagonistic behavior, due to limited effects on microbiota composition between the divergently selected lines for high and low FP [98]. It is still unclear how the gut microbiota is involved in injurious behaviors. In addition, the effects of early-life microbiota transplantation on gut microbiota composition and its function have not been well established [99]. For these reasons, we have designed and tested our hypothesis: modulation of the gut microbiota via cecal microbiota transplantation (CMT) from divergently selected inbred genetic lines (donors) would alter injurious behaviors in egg-laying chickens (recipients).

2. Genetic Lines and Study Design

In our pilot study, chicks (day-old) orally inoculated with cecal microbiota from divergently selected donors (non-aggressive or aggressive hens) altered injurious behaviors in recipients; i.e., non-aggressive donors’ recipients showed less aggressive pecking than aggressive donors’ recipients with higher brain serotonergic activities.

2.1. Unique Production, Biology, and Behavior between the Divergently Selected Inbred Lines

Two unique highly inbred white leghorn chicken lines have been continuously selected for resistance (line 63) or susceptibility (line 72) to Marek’s disease since the late 1960s [100,101,102]. This selection leads to line differences in production performance [103], neuroendocrine function [104,105,106], immunity [107,108,109,110], and behavior [111,112]. Compared to line 63 chickens, line 72 chickens have a higher number of CD4+ T cells but a lower number of CD8+ T cells [113,114] with suppressed cellular immunity [115]. In addition, the expression of cytokine (interleukin (IL)-6 and IL-18) mRNA in response to Marek’s disease virus infection is significantly different between the two inbred lines, of which line 72 chickens express higher levels of both cytokines than line 63 chickens [116], while line 63 chickens have higher gene expressions of toll-like receptor (TLR)-3, TLR-7, and IL-8 [117]. Toll-like receptors, as a class of proteins, are expressed on the membranes of various immune cells, playing a key role in the innate immune system. IL-8, as a chemoattractant cytokine, attracts and activates neutrophils in inflammatory regions via regulating the innate immune system. In addition, line differences in social stress and stress-induced aggressive behaviors have been observed; line 72 chickens have higher heterophil-to-lymphocyte (H/L) ratios (a stress indicator) with more aggressive pecks and longer durations of fights than those of line 63 chickens [102,104,112]. The differences in behaviors could be related to the line differences in serotonergic activities [105]. Line 72 chickens have lower levels of brain serotonin (5-HT) than line 63 chickens. Serotonin dysregulation has been implicated in a range of neuropsychiatric disorders in humans and various animals including chickens [118,119]. Lower levels of 5-HT have also been found in the brain of violent offenders [120,121,122]. The unique divergently selected inbred lines provide useful models for investigating gut microbiota effects on injurious behaviors in chickens. To understand the role of the cecal microbiome in regulating injurious behaviors, the following trials were conducted using cecal contents from the two inbred chicken lines.

2.2. Study Design and Results

2.2.1. Trial 1 [112]

The aim of this trial was to determine the correlations between aggressive behavior, gut microbiota, and physiological characteristics of the divergently selected laying hens (lines 63 vs. 72). The samples of blood, brain (the raphe nucleus), and cecal content were collected from ten sixty-week-old hens per line (n = 10). Monoamines of the raphe nucleus (serotonin, 5-HT; 5-hydroxyindoleacetic acid, 5-HIAA; tryptophan, TRP; epinephrine, EP; and norepinephrine, NE) were measured using high-performance liquid chromatography (HPLC). Peripheral (plasma) 5-HT and TRP, cytokines (IL-2, IL-6, IL-10, and tumor necrosis factor, TNF-α), and immunoglobulin (Ig) G were detected in duplicate using enzyme-linked immunosorbent assay (ELISA). Plasma corticosterone (CORT) concentration was measured in duplicate using radioimmunoassay (RIA). The number of peripheral white blood cells was measured and then the H/L ratio was calculated. Cecal contents were used for determining the line differences of the microbiota composition using 16S rRNA sequencing analysis, and functional predictions were performed.
The results showed that central 5-HT and TRP levels were higher in line 63 chickens compared to those of line 72 chickens (p < 0.05, Table 1A). In addition, both CORT concentrations and H/L ratios were lower in line 63 chickens (p < 0.05, Table 1B). The level of TNF-α tended to be higher in line 63 chickens (p = 0.09, Table 1C). Line differences in the cecal microbial community were also found between line 63 and line 72 chickens. Line 72 chickens had higher phylogenetic diversity than line 63 chickens, with distinct microbiota composition differences (Figure 1A,B). Faecalibacterium, Oscillibacter, Butyricicoccus, and Bacteriodes were enriched in line 63 chickens, while Clostridiales vadin BB60, Alistipes, and Mollicutes RF39 were dominant in line 72 chickens (Figure 1C,D). Like the previous findings [105], function prediction from PICRUSt2 indicated that the kynurenine pathway (KP) was enriched in line 72 chickens, while tryptophan–serotonergic activity was inherently higher in line 63 chickens. The KP of tryptophan metabolism (degraded more than 90% of absorbed dietary TRP) plays a critical role in psychiatric disorders as many kynurenine metabolites are neuroactive factors modulating neuroplasticity and/or exerting neurotoxic effects. These results suggest there is a functional linkage between the line differences in the serotonergic activity, stress response, innate immunity, and cecal microbiota populations, which provides a rationale of the hypothesis that microbiota transplantation at an early age may be a novel strategy for reducing the stress response and stress-related injurious behaviors in chickens. Based on the outcomes, trials 2 and 3 were designed and conducted (Figure 2).

2.2.2. Trial 2 [106]

The aim of this trial was to determine the effects of early-life CMT from the divergently selected inbred lines on growth, gut 5-HT, and immunity in recipient chickens. The cecal contents were randomly collected from 10 sixty-week-old hens per inbred line (donors). The collected samples were evenly pooled within the line and then diluted 1:10 with gut microbiome media. The recipients were a commercial strain, Dekalb-XL-line chickens. The oral gavage of diluted cecal microbiota was conducted once daily from day 1 to day 10, and then boosted once weekly from week 3 to week 5. Eighty-four 1-day-old male chicks were randomly assigned to 3 treatments with 7 cages and 4 chicks per cage for a 16-week trial (n = 7): CTRL (control, 0.1 mL NaCl saline), 63-CMT (0.1 mL cecal solution of line 63), and 72-CMT (0.1 mL cecal solution of line 72). The male chicks were used in this study as male chickens tend to be more aggressive than female chickens due to the hormonal differences. In weeks 5 and 16, the blood samples were collected for H/L ratios, and the levels of cytokines (IL-6, IL-10, and TNF-α), IgG, and CORT were measured using white blood cell counting, ELISA, and RIA, respectively. The spleen samples were used for mRNA expression of cytokines by RT-qPCR, and the ileal samples—two 3.5 cm sections (near the diverticulum)—were collected for histomorphological analysis using a routine hematoxylin and eosin procedure. Gut serotonergic activity (TRP, 5-HT, 5-HIAA, and 5-HIAA/5-HT ratio) and secretory (s) IgA were analyzed using HPLC and ELISA, respectively. The body weight was also collected for calculating the relative weight of the adrenal gland.
The results showed that compared to 72-CMT chickens, 63-CMT chickens had a lower body weight and ileal villus/crypt ratio among the treatments in week 5 (Figure 3 and Figure S1). In addition, 63-CMT chickens had an improved stress adaptive capacity: lower H/L ratios, together with a tendency of a lower relative adrenal gland weight in week 16 (Table 2). 63-CMT chickens also had higher plasma levels of IL-10, with lower levels of plasma natural IgG, with a tendency of lower levels of IL-6 in week 16 (Table 3). In contrast, 72-CMT chickens had a lower concentration of ileal mucosal sIgA in week 5 with a tendency for a higher mRNA abundance of splenic IL-6 and TNF-α in week 16 (Table 4). Furthermore, 63-CMT chickens tended to have the highest 5-HT concentrations with the highest serotonergic turnover in the ileum in week 5 (Figure 4). These results indicate that early-postnatal CMT from the different donors (lines) was associated with the different patterns of growth and health status through regulating the ileal morphological structures, gut-derived serotonergic activity, peripheral cytokines, and antibody production, as well as stress responses in recipient chickens. The findings confirm our hypothesis that transferring cecal contents at an early age has unique line effects, including growth, immunity, and gut neurotransmitter synthesis with a long-lasting effect. The current findings may also indicate that the gut microbial function is without donor-recipient genetic effects (i.e., with the line’s unique biologic characteristics being transferred from the selected inbred donors to the third commercial recipient line regardless of the lines’ genetic backgrounds).

2.2.3. Trial 3 [123]

The aim of this trial was to determine the effects of early-life CMT from the divergently selected inbred chicken lines (donors) on cecal microbiota profile, brain monoamines, aggression, and their correlations in recipient chickens. The samples of the brain (the hypothalamus) and cecal contents of recipients were collected. The monoamines (5-HT, EP, NE, and DA) of the hypothalamus were measured in triplicate using HPLC, and cecal samples were analyzed using 16S rRNA gene sequencing. The aggressive behaviors were observed using both home-cage video analysis and paired behavioral tests based on the previously developed definitions [124,125,126]. The paired test is a routine method used for analyzing aggression-related social ranking as well as fear and anxiety in chickens [127,128,129]. Its rationale and mechanisms are similar to the resident–intruder test used in rodents, a standardized test for detecting social-stress-induced aggression and related violence [130].
Data indicated that compared to 72-CMT recipients, 63-CMT recipients showed less aggressive behaviors (Figure 5) with a higher serotonergic activity, evidenced by higher concentrations of 5-HT and 5-HIAA (Table 5) in the hypothalamus in week 5 with a tendency for higher concentrations of TRP in week 16. Tryptophan can pass the blood–brain barrier and is the sole precursor of 5-HT [51], which may indirectly indicate how to reduce aggressive behaviors in the recipient chickens via activating the brain serotonergic system. Through 16S rRNA gene sequence analysis, we observed that CMT-induced microbiota changes, that is, a distinct microbial community diversity, were observed between 63-CMT and 72-CMT recipients. 72-CMT recipient chickens had a higher phylogenetic diversity than 63-CMT recipient chickens in weeks 5 and 16 (Figure 6A). Cecal microbiota transplantation also induced changes in microbial community structures (Unweighted UniFrac) among treatments in week 5 but not in week 16 (Figure 6B). Compared to 63-CMT chickens, 72-CMT chickens had enriched ASVs belonging to 14 genera, including Akkermansia, Anaeroplasma, Ruminococcaceae UCG-008, Faecalibacterium, Blautia, Dielma, GCA-900066225, Merdibacter, and CAG-56, while ASVs belonging to 5 genera were more abundant in 63-CMT recipients including Ruminococcaceae UCG-005, Ruminococcaceae UCG-014, Lachnospiraceae, and Fournierella. In week 16, compared to 63-CMT recipients, ASVs belonging to 5 genera (including Bacillus, Escherichia-Shigella, Lachnospiraceae, and Bacteroides) were more abundant in the 72-CMT recipients, while ASVs belonging to 6 genera were more abundant in 63-CMT recipients (including Ruminococcaceae NK4A214 group, Ruminococcaceae, and Eubacterium coprostanoligenes group) (Figure 6C). The results suggest that CMT at an early age affects the development of the gut microbiota composition and reduces aggressive behaviors in recipient chickens via regulating the activities of the brain serotonergic system through the gut–brain axis.

3. Cecal Microbiota Transplantation, Social Stress, and Injurious Behavior in Chickens

3.1. Stress and Gut Microbiota

Stress is a natural biological (physical and mental) response to internal and external challenges in living organisms, including chickens. Normally, it prompts chickens’ ability to adapt to their rearing environments, while abnormally, an overload of stress challenge (too much exposure to a stressor or combined stressors causing a long-term activation of the stress response systems) reduces gut microbiota diversity, composition, or both [131]. The gut microbiota is functionally like an endocrine organ, releasing numerous bioactive factors to activate the HPA and SAM stress systems in response to stimulations, consequently affecting host physiological and behavioral homeostasis via the bidirectional communication of the MGB and MGI axes [63,64]. Healthy intestinal microbial communities and functions are essential for animals to fit their living environments [132,133]. The intestinal microbial community has been named the “social or behavioral immune system” linked to the microbiota–gut–brain–immune axis [134] based on the two reciprocal themes: (1) that gut microbiota influences host social behavior and (2) that social behavior and social structure shape the composition of the gut microbiota across individuals [135]. Based on these theories, environmental factors causing changes in the gut microbiome are linked to stress-induced neurobehavioral disorders including aggression and related damaging behaviors [136,137]. In addition, the differences in gut microbiota composition and/or diversity are related to personality traits [15,138], temperament [139,140], and sociability [87,141] in humans and various social animals, including chickens.
Numerous psychological (an emotion and/or mental overstimulation) and/or physical (environmental conditions) stressors reduce gut microbiota diversity and/or alter microbiome composition by (1) disrupting the community stability of commensal bacterial populations, often accompanied by reduced beneficial bacteria and increased pathogens (causing a chronic low-grade inflammation); (2) increasing the survival translocation of pathogens and releasing virulence factors; (3) disrupting absorption of nutrients and minerals (metabolic disorders); (4) disrupting microbial neuroendocrine functions (alterations in synthesis of several signaling molecules and neurochemicals including 5-HT in the GIT); (5) disrupting the gut epithelial barrier, thereby increasing intestinal permeability and releasing certain bacteria, bacterial antigens, and metabolites (leaky gut), resulting in both intestinal and systemic immune reactions; and (6) damaging epithelial cells, producing free radicals and reducing antioxidant capacity (oxidative stress) [142,143,144]. These changes in the gut microbiota with a chronic low-grade inflammation profoundly influence host health and behavioral homeostasis via the MGB and MGI axes [58,145]. Treatments aimed at restoring normal gut microbiota composition and homeostasis have become effective methods to prevent and/or reduce various stress-induced neuropsychiatric disorders [146,147].

3.2. Possible Pathophysiological Mechanisms Underlying Injurious Behaviors in Chickens

In mammals, chronic stress is a major risk factor in neuropsychiatric disorders [148]. Social stress induces numerous microbiota-derived neurochemicals (neuromodulators) to enter the blood stream and influence brain function, especially the functions of both the HPA and SAM axes [149,150], which affects the development and balance of emotional and mental behaviors. Alterations in neuroendocrine homeostasis, i.e., CORT and catecholamines (such as EP and NE) levels, have been identified as the final common pathways in controlling animal behavior and pathophysiological status [151]. Animals raised in a germ-free (GF) environment expressing an exaggerated HPA response to psychological stressors could be normalized with certain bacterial probiotic species, such as Bifidobacterium infantis [152,153] and Bacillus licheniformis [154]. The animals treated with probiotics had a blunted HPA response [155]. Similarly, FP in chickens is influenced by dysregulation of the gut microbiome, which consequently affects neurotransmitter and immune homeostasis [27,94,95]. Current studies have evidenced that changing prenatal and early postnatal brain developments are involved in the development of injurious behaviors in laying hens [156] and other farm animals [157]. Our current studies have evidenced that early-life CMT induced different levels of aggressive behavior in the male recipients, which is corrected with each donor line’s behaviors. The results indicate that transferred donors’ cecal microbiota uniquely modifies the serotonergic activity, stress response, innate immunity, and cecal microbiota populations in recipients through the MGB and MGI axes. The underlying mechanisms, such as the responsible individual bacterium (or bacteria), the released neuromodulators and/or metabolites, as well as the involved pathways, will be examined in upcoming studies.

3.3. Physiological Mechanisms of Modulation of Intestinal Microbiota to Regulate Social Stress and Related Abnormal Behaviors

A healthy intestinal microbial community plays a critical role in regulating stress responses of the HPA and SAM axes to maintain host behavioral and physiological functions to fit their living environments [132]. Accumulating studies from various animal models in gut microbiota investigations, such as GF (complete absence of microbial exposure) animals, SPF (specific pathogen-free) animals, antibiotic-treated (broad-spectrum antibiotic cocktails) animals, and animals exposed to pathogenic bacterial infections, suggest that the gut microbiota plays an important role in the regulation of anxiety, mood, and cognition, indicating the possibility of using probiotics to modify the gut microbiota to control impulsive and compulsive behaviors in patients with neuropsychiatric disorders [158,159,160,161]. Like mammals, the gut microbiome plays a critical role in poultry health and welfare [119,162,163]. Laying hens showing high or low FP have different gut microbial populations [27,94,95,164] and metabolite profiles [96,97]. Therefore, the gut microbiome represents a novel therapeutic target for stress-induced mental and mood disorders in humans and injurious behaviors in chickens.
Probiotics are commensal bacteria that offer potential health benefits to the host, including the allostatic load (cumulating chronic stress effects on the body), when administered in adequate amounts. Generally, probiotics may aid animals in adapting to their ambient environments and protect against pathogens by (1) altering the microbiota profile in favor of beneficial bacteria to prevent the growth of pathogens and compete with enteric pathogens for the limited availability of nutrient and attachment sites; (2) producing bacteriocins (including bacteriostatic and bactericidal substances) and short-chain fatty acids against pathogens to regulate the activity of intestinal digestive enzymes and energy homeostasis and increase mineral solubility; (3) modulating host immune and inflammatory responses to reduce oxidative stress, inflammation, and cell injury; (4) restoring/strengthening the intestinal barrier integrity, which prevents pathogens and toxic substances from crossing the mucosal epithelium; (5) stimulating the neuroendocrine system and attenuating stress-induced disorders of the HPA and/or SAM axes via the MGB and MGI axes; and/or (6) inducing epithelial heat shock proteins to protect cells from oxidative damage [165,166,167,168,169,170]. Both human and rodent studies indicated that probiotics reduce chronic-psychological-stress-induced abnormal brain activity and related cognitive dysfunctions by lowering plasma CORT and adrenocorticotropic hormone levels, restoring hippocampal 5-HT and NE levels, and normalizing immunity with low plasma levels of TNF-α but high levels of IL-10, an anti-inflammatory cytokine [171,172,173]. Several probiotics, as psychobiotics, for example, Bifidobacterium and Lactobacillus, deliver mental health benefits with neurobehavioral effects, which have been used in humans for improving cognitive function and for preventing and treating patients with behavioral impairment in neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease, and in diseases with neuropsychiatric disorders, such as anxiety, depression, and impulsively and compulsively disrupted social behavior [75,77,78,79,83,84]. Based on findings, targeting the gut microbiota has been recognized as a novel therapeutic option for patients with neuropsychiatric disorders [63,73,74]. Current studies have evidenced that the influence of the gut microbiota on the host behavior as seen in mammals is shared in chickens [173]. For example, dietary supplements of probiotics-based Bacillus amyloliquefaciens reduce distress calls and aggressive behavior in turkey poults [174], and Lactobacillus rhamnosus [175,176] and Bacillus subtilis [177] decrease stress-induced FP in adult hens by restoring the gut microbiota and 5-HT metabolism [70]. However, the evidence for probiotic benefits is mixed, proposing that the use of live commensals coming directly from a healthy gut may be more effective than probiotics.
Fecal microbiota transplantation has recently become a novel method for modulating the gut microbiota in gastrointestinal disorders such as inflammatory bowel syndrome and CDI [178,179], and non-gastrointestinal diseases including neuropsychiatric disorders [180,181]. Fecal microbiota transplantation is a method of directly restoring healthy gut bacteria by transferring stool from a healthy donor. Stool contains thousands of microorganisms and a vast number of metabolites and has been recognized as a rapid and effective method to reshape the intestinal microbiota and metabolic profiles in humans and animals [182,183]. For example, the gut microbiota of recipients from stressed donors mimics the effects of stress on control animals, which could be reversed by transferring microbiota from unstressed animals [184,185]. Studies in CDI patients revealed that the diversity of gut microbiota is increased following FMT, which is critical for defense against pathogens via colonization resistance. Clinically, a single dose can have long-lasting effects [186,187,188]. However, recent studies indicate that a fecal sample is not reliable in mapping the complete cecal microbiome and cannot be used to monitor the shifts and changes in cecal content in chickens [189,190,191].
Taken together, in humans and rodents, microbial colonization impacts brain development in early life, with long-lasting effects on adult behavior. Fecal microbiota transplantation and probiotics repair the social-stress-induced disturbance of microbial functions and attenuate the stress-induced responses of the HPA and/or SAM axes by protecting neuronal plasticity at the hypothalamic level as well as promoting neurogenesis in the hippocampus. Fecal microbiota transplantation restores the negative feedback of the stress systems to regulate animal health and behavior, providing novel insights into understanding how the gut microbiota community prevents abnormal behavior in patients with psychological disorders. We hypothesized that similar cellular mechanisms may be manifested in CMT recipient chickens, because chickens and mammals share a similarity in the interactions between the microbiome and the neuroendocrine systems, generally named microbial endocrinology [192,193,194]. This hypothesis has been tested and evidenced in our recent studies.

3.4. Cecal Microbiota Transplantation and Injurious Behavior in Chickens

Early life (immediate post-hatch) in chickens is a critical window of time causing enduring effects on the development of the intestinal microbiome and related brain functions and behavior in later life. Although microbial complexity considerably increases in the cecum with age [157], modulation of the structure and function of the cecal microbiome during early life alters neurophysiology in adolescence [195]. In chickens, the avian cecum plays a vitally important role in maintaining pathophysiological homeostasis, especially during periods of stress [196,197,198,199]. With up to 1011 cells per gram of content, the cecum has the greatest bacterial biodiversity (bacterial diversity, richness, and species composition) along the chicken GIT [200,201,202]. As a multi-purpose organ, it has a complex motility, pushing contents in two directions (a two-component system): the cloaca (excreting as cecal drop) and the ileum (providing bacteria (for bacterial proliferation and colonization)) involved in the bird’s biological homeostasis [197,203,204,205]. The cecum with its high level of diversity maintains intestinal microbial stability in responding to various stressors [206] and determines colonization resistance against invading pathogens [207]. As the bird’s primary fermentative organ, the cecum possesses higher levels of DNA replicative viability than feces [208]. A balanced cecal microbiota diversity and composition have been used as an indicator of growth and health in poultry [209,210,211]. However, unlike mammals, in a commercial production setting, microbial contact is completely interrupted between domesticated parents and chicks. Various technologies have been developed for the modification of gut microbiota diversity and composition and related functions, including CMT, in chickens.
The effects of early-life CMT on the development of the gut microbiota in recipient chickens with long-lasting effects have been previously investigated. Franco et al. reported that broiler chicks (recipients) that received cecal contents from organic hens or industry-raised broilers (donors) by oral application on day 1 had distinctly colonized bacterial microbiota profiles, which was similar to the cecal microbiota profiles of the donors, respectively [212]. The differences between the recipient broilers had been maintained from day 7 to day 42 (the end of this study). The results indicate that transferred microbiota can persistently colonize the newly hatched broilers. In addition, early intervention with cecal fermentation broth from donor broilers (180 days old) regulates the colonization and development of gut microbial function in newly hatched broiler chicks (recipients), increasing beneficial bacteria and the concentration of short-chain fatty acids (SCFAs), while reducing the abundance of pathogenic bacteria [213]. In another study, cecal contents collected from ISA Brown chickens or hens (donors) at 1, 3, 16, 28, and 42 weeks of age were orally applied to newly hatched broiler chicks (recipients) [214]. Its results showed that the cecal proteome of recipient chicks was correlated to the composition of the donors’ microbiome following a single inoculation on the day of hatch, with a long-lasting effect, up to 45 days of age (an entire broiler production period). Taken together, early inoculation with cecal microbiota represents a novel method for modulating the host microbiome to improve production and reduce susceptibility to infection in chickens.
In the current studies, CMT from the divergently selected inbred donor lines has been evidenced functionally to reduce or inhibit the stress response and related aggression and damage pecking in recipient chickens of a commercial strain. These findings further support the theory that the exhibition of injurious behaviors is a stress-induced neuropsychological disorder in chickens, which is comparable to human psychopathological disorders [215,216]. Stress-associated gut dysbiosis and low-grade chronic inflammation are common traits of these disorders. For group-living chickens as well as other social animals, individuals share microbes and interact around environments and resources, by which the gut microbiota may have considerable consequences for host social interactions, such as the social ranking of individual animals [217,218]. For laying hens, like other social animals, the development of injurious behaviors may therefore be a phenotypic behavioral consequence of an imbalanced gut microbiota composition and related dysregulation of the communication between the gut and brain [204]. Birds with a higher propensity to perform injurious behaviors have distinct microbiota profiles compared to their non-pecking counterparts [27,112]. Similarly, the microbiota differing between the selected inbred lines (line 63 vs. line 72) exhibit distinct phenotypes [112], and CMT may be a method with the potential to control and replicate the role of the gut microbial community after a single passage of transplanted cecal content. This hypothesis will be tested in upcoming studies.
Major microbiota colonization of the intestine occurs in post-hatched chicks. CMT in early life (day-old chicks) may have great protective effects against stress-induced physiological and behavioral changes [219,220]. The current study showed that recipient chickens (63-CMT compared to 72-CMT) had different levels of aggression and related damaging behaviors, which was correlated with the degree of injurious behaviors of donors [123]. The early postnatal period is a vital window for birds as well as mammals to be colonized with the microbiome [213], whereby early-life CMT profoundly influences brain development and intestinal microbiota composition and diversity [221] with a long-lasting impact on gut–brain neural circuit development and its responses to stressful episodes [222,223]. However, inconsistent results of CMT-induced intestinal microbiota modulation have been reported across studies. Early-life homologous (within line) microbiota transplantation (a pooled donor’s ileum, ceca, and colon contents) increases activation in both selected high- and low-FP recipients, with limited effects on their microbiota composition, stress response, and FP [28]. It is still unclear how FP arises as a consequence of dysregulated communication between the gut and the brain. A recent study also reported that gut microbial composition (from the digesta and mucosa of the ileum and cecum) and predicted functions were not associated with FP and antagonistic behavior in laying hens [33]. Therefore, given the inconsistent results, there is a critical need to further identify the biofunctions of cecal microbiota in controlling injurious behaviors in laying hens via CMT from the divergently selected non-aggressive and aggressive lines. Taken together, the obtained results may potentially influence the common procedures used in controlling aggression and related injurious behaviors in chickens as well as other species of farm animals, such as the dehorning of calves in beef and dairy operations [224,225] and teeth clipping or tail docking in swine operations [226,227]. Our work may also have implications for human medicine, providing information for developing next-generation psychobiotics [228,229] and impacting human mental health; currently, 1 in 6 U.S. youth aged 6–17 and 1 in 5 U.S. adults experience mental health disorders each year [230].

4. Conclusions and Perspectives

The current results show that differences in behavior, serotonergic activity, stress response, innate immunity, and cecal microbiota populations between the two divergently selected inbred genetic lines (donors, line 63 vs. line 72) can be transferred to other chicken lines (recipients) at an early age (day-old in this study) with long-lasting effects on growth, behavior, and biological functions. The data suggest that the CMT effects are independent of genetic differences between the donors and recipients. The outcomes provide new insights into understanding the underlying mechanisms of the MGB and MGI axes in regulating abnormal behaviors and offer a tractable strategy for reducing social stress and stress-associated injurious behaviors and improving welfare in egg-laying chickens. The roles of individual cecal bacterial members (as the optimal next-generation psychobiotics), the released bioactive factors (as the next-generation agents), and the related biological processes underlying social stress and injurious behaviors in chickens will be examined in the following studies.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/microorganisms12030471/s1. Figure S1: The examples of the morphological changes of the villus height and crypt depth in the ileum of recipient chickens at (A) week 5 and (B) week 16. Magnification: × 100. Villus height (VH); Crypt depth (CD).

Funding

This research received no external funding.

Acknowledgments

We sincerely thank the farm staff of Purdue Poultry Animal Sciences Research Center, and the technicians of the USDA-ARS, Livestock Behavior Research Unit, for their assistance with this project. Mention of trade names or commercial products in this article is solely for the purpose of providing specific information and does not imply recommendation or endorsement by the USDA. The USDA is an equal opportunity provider and employer. The data presented in this review have been previously published in our earlier papers, included here to enhance the audience’s comprehension. Additionally, proper citation of the data is provided within this review.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-HIAA, 5-hydroxuindoleacetic acid; 5-HT, serotonin; 63-CMT, a recipient chicken with cecal content solution of line 63; 72-CMT, a recipient chicken with cecal content solution of line 72; ASV, amplicon sequence variant; BW, body weight; BT, beak trimming; CMT, cecal microbiota transplantation; CD, crypt depth; CDI, C. difficile infection; CORT, corticosterone; DA, dopamine; ELISA, enzyme-linked immunosorbent assay; EP, epinephrine; FMT, fecal microbiota transplantation; FP, feather pecking; GIT, gastrointestinal tract; GF, germ free; HPA, hypothalamic–pituitary–adrenal axis; H/L, heterophil/lymphocyte ratio; HPLC, high-performance liquid chromatography; Ig, immunoglobulin; IL, interleukin; KP, kynurenine pathway; MGB, microbiota–gut–brain axis; MGI, microbiota–gut–immune axis; NE, epinephrine; SAM, sympathetic–adrenal–medullary axis; sIgA, secretory IgA; SCFA, short-chain fatty acids; SFP, severe feather pecking; SPF, specific pathogen-free; TLR, toll-like receptor; TNF-α, tumor necrosis factor alpha; TRP, tryptophan; VH, villus height.

References

  1. Henchion, M.; Moloney, A.P.; Hyland, J.; Zimmermann, J.; McCarthy, S. Review: Trends for meat, milk and egg consumption for the next decades and the role played by livestock systems in the global production of proteins. Animal 2021, 15, 100287. [Google Scholar] [CrossRef] [PubMed]
  2. USDA ERS (Economic Research Service, U.S. Department of Agriculture). Poultry and Eggs, Market Outlook. Available online: https://www.ers.usda.gov/topics/animal-products/poultry-eggs/market-outlook (accessed on 30 November 2023).
  3. Hagelso, A.M.; Krohn, C.C. Quantifing social behavior of the individual. In Animal Genetic Resources for Adaptation to More Extensive Production Systems; Hagelso, A.M., Ed.; Commission of the European Communities: Foulum, Denmark, 1993. [Google Scholar]
  4. Rothschild, J. Ethical considerations of gene editing and genetic selection. J. Gen. Fam. Med. 2020, 21, 37–47. [Google Scholar] [CrossRef]
  5. Muir, W.M.; Cheng, H.W.; Coney, C. New selection methods for layer performance and potential impacts on behavior and management. In Proceedings of the XIVth European Poultry Conference, Stavanger, Norway, 23–27 June 2014. [Google Scholar]
  6. Baldauf, S.; Engqvist, L.; Weissing, F. Diversifying evolution of competitiveness. Nat. Commun. 2014, 5, 5233. [Google Scholar] [CrossRef]
  7. Bernhardt, J.R.; Kratina, P.; Pereira, A.L.; Tamminen, M.; Thomas, M.K.; Narwani, A. The evolution of competitive ability for essential resources. Philos. Trans. R. Soc. 2020, B375, 20190247. [Google Scholar] [CrossRef] [PubMed]
  8. Grether, G.F.; Anderson, C.N.; Drury, J.P.; Kirschel, A.N.; Losin, N.; Okamoto, K.; Peiman, K.S. The evolutionary consequences of interspecific aggression. Ann. N. Y. Acad. Sci. 2013, 1289, 48–68. [Google Scholar] [CrossRef] [PubMed]
  9. Hocking, P.M. Unexpected consequences of genetic selection in broilers and turkeys: Problems and solutions. Br. Poult. Sci. 2014, 55, 1–12. [Google Scholar] [CrossRef] [PubMed]
  10. Drury, J.P.; Cowen, M.C.; Grether, G.F. Competition and Hybridization Drive Interspecific Territoriality in Birds. Proc. Natl. Acad. Sci. USA 2020, 117, 12923–12930. [Google Scholar] [CrossRef] [PubMed]
  11. Muir, W.M. Group Selection for Adaptation to Multiple-Hen Cages: Selection Program and Direct Responses. Poult. Sci. 1996, 75, 447–458. [Google Scholar] [CrossRef]
  12. Biro, P.A.; Stamps, J.A. Are animal personality traits linked to life-history productivity? Trends Ecol. Evol. 2008, 23, 361–368. [Google Scholar] [CrossRef]
  13. Senner, N.R.; Conklin, J.R.; Piersma, T. An ontogenetic perspective on individual differences. Proc. Biol. Sci. 2015, 282, 20151050. [Google Scholar] [CrossRef]
  14. Moore, M.P.; Martin, R.A. On the evolution of carry-over effects. J. Anim. Ecol. 2019, 88, 1832–1844. [Google Scholar] [CrossRef] [PubMed]
  15. Chunduri, A.; Reddy, S.D.M.; Jahanavi, M.; Reddy, C.N. Gut–Brain Axis, Neurodegeneration and Mental Health: A Personalized Medicine Perspective. Indian J. Microbiol. 2022, 62, 505–515. [Google Scholar] [CrossRef] [PubMed]
  16. Mench, J.A.; Duncan, I.J. Poultry welfare in North America: Opportunities and challenges. Poult. Sci. 1998, 77, 1763–1765. [Google Scholar] [CrossRef]
  17. Cheng, H.-W. Animal Welfare: Should We Change Housing to Better Accommodate the Animal or Change the Animal to Accommodate the Housing? CABI Rev. 2007, 2, 14. [Google Scholar] [CrossRef]
  18. Lin, E.-J.D.; Sun, M.; Choi, E.Y.; Magee, D.; Stets, C.W.; During, M.J. Social Overcrowding as a Chronic Stress Model That Increases Adiposity in Mice. Psychoneuroendocrinology 2015, 51, 318–330. [Google Scholar] [CrossRef]
  19. Banich, M.T.; Floresco, S. Reward systems, cognition, and emotion: Introduction to the special issue. Cogn. Affect. Behav. Neurosci. 2019, 19, 409–414. [Google Scholar] [CrossRef]
  20. Kern, E.M.A.; Robinson, D.; Gass, E.; Godwin, J.; Langerhans, R.B. Correlated evolution of personality, morphology and performance. Anim. Behav. 2016, 117, 79–86. [Google Scholar] [CrossRef]
  21. De Boer, S.F. Animal models of excessive aggression: Implications for human aggression and violence. Curr. Opin. Psychol. 2018, 19, 81–87. [Google Scholar] [CrossRef]
  22. Flanigan, M.E.; Russo, S.J. Recent advances in the study of aggression. Neuropsychopharmacology 2019, 44, 241–244. [Google Scholar] [CrossRef]
  23. Kleszcz, A.; Cholewińska, P.; Front, G.; Pacoń, J.; Bodkowski, R.; Janczak, M.; Dorobisz, T. Review on Selected Aggression Causes and the Role of Neurocognitive Science in the Diagnosis. Animals 2022, 12, 281. [Google Scholar] [CrossRef]
  24. Lay, D.C., Jr.; Fulton, R.M.; Hester, P.Y.; Karcher, D.M.; Kjaer, J.B.; Mench, J.A.; Mullens, B.A.; Newberry, R.C.; Nicol, C.J.; O’Sullivan, N.P.; et al. Hen welfare in different housing systems. Poult. Sci. 2011, 90, 278–294. [Google Scholar] [CrossRef] [PubMed]
  25. van Staaveren, N.; Harlander, A. Cause and Prevention of Injurious Pecking in Chickens. In Understanding the Behaviour and Improving the Welfare of Chickens; Burleigh Dodds Science Publishing: Cambridge, UK, 2020; pp. 509–566. [Google Scholar]
  26. Meuser, V.; Weinhold, L.; Hillemacher, S.; Tiemann, I. Welfare-Related Behaviors in Chickens: Characterization of Fear and Exploration in Local and Commercial Chicken Strains. Animals 2021, 11, 679. [Google Scholar] [CrossRef] [PubMed]
  27. Van der Eijk, J.A.; de Vries, H.; Kjaer, J.B.; Naguib, M.; Kemp, B.; Smidt, H.; Rodenburg, T.B.; Lammers, A. Differences in gut microbiota composition of laying hen lines divergently selected on feather pecking. Poult. Sci. 2019, 98, 7009–7021. [Google Scholar] [CrossRef]
  28. Van der Eijk, J.A.; Rodenburg, T.B.; de Vries, H.; Kjaer, J.B.; Smidt, H.; Naguib, M.; Kemp, B.; Lammers, A. Early-life microbiota transplantation affects behavioural responses, serotonin and immune characteristics in chicken lines divergently selected on feather pecking. Sci. Rep. 2020, 10, 2750. [Google Scholar] [CrossRef] [PubMed]
  29. Iffland, H.; Wellmann, R.; Preuß, S.; Tetens, J.; Bessei, W.; Piepho, H.P.; Bennewitz, J. A novel model to explain extreme feather pecking behavior in laying hens. Behav. Genet. 2020, 50, 41–50. [Google Scholar] [CrossRef] [PubMed]
  30. Cronin, G.; Hopcroft, R.; Groves, P.; Hall, E.; Phalen, D.; Hemsworth, P. Why Did Severe Feather Pecking and Cannibalism Outbreaks Occur? An Unintended Case Study While Investigating the Effects of Forage and Stress on Pullets during Rearing. Poult. Sci. 2018, 97, 1484–1502. [Google Scholar] [CrossRef]
  31. Ellen, E.D.; Van Der Sluis, M.; Siegford, J.; Guzhva, O.; Toscano, M.J.; Bennewitz, J.; Van Der Zande, L.E.; Van Der Eijk, J.A.; de Haas, E.N.; Norton, T.; et al. Review of sensor technologies in animal breeding: Phenotyping behaviors of laying hens to select against feather pecking. Animals 2019, 9, 108. [Google Scholar] [CrossRef]
  32. Falker-Gieske, C.; Mott, A.; Preuß, S.; Franzenburg, S.; Bessei, W.; Bennewitz, J.; Tetens, J. Analysis of the brain transcriptome in lines of laying hens divergently selected for feather pecking. BMC Genom. 2020, 21, 595. [Google Scholar] [CrossRef]
  33. Falker-Gieske, C.; Iffland, H.; Preuß, S.; Bessei, W.; Drögemüller, C.; Bennewitz, J.; Tetens, J. Meta-analyses of genome wide association studies in lines of laying hens divergently selected for feather pecking using imputed sequence level genotypes. BMC Genet. 2020, 21, 114. [Google Scholar] [CrossRef]
  34. Baker, P.E.; Nicol, C.J.; Weeks, C.A. The effect of hard pecking enrichment during rear on feather cover, feather pecking behaviour and beak length in beak-trimmed and intact-beak laying hen pullets. Animals 2022, 12, 674. [Google Scholar] [CrossRef]
  35. BFREPA (the British Free Range Egg Producers Association). Egg Industry Organizations Have Joined Forces to Fund Vital Research into the Latest Beak Treatment Technique. 2006. Available online: http://www.theranger.co.uk (accessed on 30 November 2023).
  36. Poultrysite. Pecking—The Unanswered Question. 2010. Available online: https://www.farminguk.com/news/pecking-the-unanswered-questions_432.html (accessed on 30 November 2023).
  37. Piepho, H.P.; Lutz, V.; Kjaer, J.B.; Grashorn, M.; Bennewitz, J.; Bessei, W. The presence of extreme feather peckers in groups of laying hens. Animal 2017, 11, 500–506. [Google Scholar] [CrossRef]
  38. RSPCA (The Royal Society for the Prevention of Cruelty to Animals). How Can Feather Pecking Be Managed in Cage-Free Layer Hen Systems? 2023. Available online: https://kb.rspca.org.au/knowledge-base/how-can-feather-pecking-be-managed-in-cage-free-layer-hen-systems/ (accessed on 30 November 2023).
  39. Cheng, H.W. Morphopathological changes and pain in beak trimming laying hens. World’s Poult. Sci. J. 2005, 62, 41–52. [Google Scholar] [CrossRef]
  40. Mikoni, N.A.; Guzman, D.S.; Fausak, E.; Paul-Murphy, J. Recognition and assessment of pain-related behaviors in avian species: An integrative review. J. Avian Med. Surg. 2022, 36, 153–172. [Google Scholar] [CrossRef]
  41. NASS (The National Agricultural Statistics Service). Chickens and Eggs. 2022. Available online: https://www.nass.usda.gov/Publications/Todays_Reports/reports/ckeg0422.pdf (accessed on 30 November 2023).
  42. Shahbandeh, M. Total Number of Laying Hens in the U.S. 2000–2022. Statista. 2023. Available online: https://www.statista.com/statistics/195823/total-number-of-laying-hens-in-the-us-since-2000/ (accessed on 30 November 2023).
  43. Iqbal, A.; Moss, A.F. Review: Key tweaks to the chicken’s beak: The versatile use of the beak by avian species and potential approaches for improvements in poultry production. Animal 2021, 15, 100119. [Google Scholar] [CrossRef]
  44. Yamauchi, Y.; Yoshida, S.; Matsuyama, H.; Obi, T.; Takase, K. Morphologically abnormal beaks observed in chickens that were beak-trimmed at young ages. J. Vet. Med. Sci. 2017, 79, 1466–1471. [Google Scholar] [CrossRef]
  45. Coton, J.; Guinebretière, M.; Guesdon, V.; Chiron, G.; Mindus, C.; Laravoire, A.; Pauthier, G.; Balaine, L.; Descamps, M.; Bignon, L.; et al. Feather pecking in laying hens housed in free-range or furnished-cage systems on French farms. Br. Poult. Sci. 2019, 60, 617–627. [Google Scholar] [CrossRef]
  46. Elson, A. Laying Hens Beaks: To Trim or Not to Trim. Livestock Knowledge Transfer, a DEFRA Initiative Operated by ADAS/IGER/University of Bristol. Poultry 2001. Available online: http://www.agrowebcee.net/fileadmin/content/faw/doc/reports2/BEAK_TRIMMING_REVIEW.pdf (accessed on 30 November 2023).
  47. Rudkin, C. Feather pecking and foraging uncorrelated—The redirection hypothesis revisited. Br. Poult. Sci. 2022, 63, 265–273. [Google Scholar] [CrossRef] [PubMed]
  48. Kops, M.S.; de Haas, E.N.; Rodenburg, T.B.; Ellen, E.D.; Korte-Bouws, G.A.; Olivier, B.; Güntürkün, O.; Bolhuis, J.E.; Korte, S.M. Effects of feather pecking phenotype (severe feather peckers, victims and non-peckers) on serotonergic and dopaminergic activity in four brain areas of laying hens (Gallus gallus domesticus). Physiol. Behav. 2013, 120, 77–82. [Google Scholar] [CrossRef]
  49. García-Cabrerizo, R.; Carbia, C.; O’ Riordan, K.J.; Schellekens, H.; Cryan, J.F. Microbiota-gut-brain axis as a regulator of reward processes. J. Neurochem. 2021, 157, 1495–1524. [Google Scholar] [CrossRef] [PubMed]
  50. Kaur, A.; Chen, T.; Green, S.J.; Mutlu, E.; Martin, B.R.; Rumpagaporn, P.; Patterson, J.A.; Keshavarzian, A.; Hamaker, B.R. Physical inaccessibility of a resistant starch shifts mouse gut microbiota to butyrogenic firmicutes. Mol. Nutr. Food Res. 2019, 63, e1801012. [Google Scholar] [CrossRef] [PubMed]
  51. Gao, K.; Mu, C.-L.; Farzi, A.; Zhu, W.-Y. Tryptophan metabolism: A link between the gut microbiota and brain. Adv. Nutr. 2020, 11, 709–723. [Google Scholar] [CrossRef] [PubMed]
  52. Rea, K.; Dinan, T.G.; Cryan, J.F. The microbiome: A key regulator of stress and neuroinflammation. Neurobiol. Stress 2016, 4, 23–33. [Google Scholar] [CrossRef] [PubMed]
  53. Gwak, M.G.; Chang, S.Y. Gut-Brain Connection: Microbiome, Gut Barrier, and Environmental Sensors. Immune Netw. 2021, 21, e20. [Google Scholar] [CrossRef]
  54. Barton, J.R.; Londregan, A.K.; Alexander, T.D.; Entezar, A.A.; Covarrubias, M.; Waldman, S.A. Enteroendocrine cell regulation of the gut-brain axis. Front. Neurosci. 2023, 17, 1272955. [Google Scholar] [CrossRef]
  55. Yarandi, S.S.; Peterson, D.A.; Treisman, G.J.; Moran, T.H.; Pasricha, P.J. Modulatory effects of gut microbiota on the central nervous system: How gut could play a role in neuropsychiatric health and Diseases. J. Neurogastroenterol. Motil. 2016, 22, 201–212. [Google Scholar] [CrossRef]
  56. Dicks, L.M.T. Gut bacteria and neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef] [PubMed]
  57. Kim, G.H.; Shim, J.O. Gut microbiota affects brain development and behavior. Clin. Exp. Pediatr. 2022, 66, 274–280. [Google Scholar] [CrossRef]
  58. Homer, B.; Judd, J.; Mohammadi Dehcheshmeh, M.; Ebrahimie, E.; Trott, D.J. Gut microbiota and behavioural issues in production, performance, and companion animals: A systematic review. Animals 2023, 13, 1458. [Google Scholar] [CrossRef]
  59. Tan, H.E. The microbiota-gut-brain axis in stress and depression. Front. Neurosci. 2023, 17, 1151478. [Google Scholar] [CrossRef]
  60. Yoshikawa, S.; Taniguchi, K.; Sawamura, H.; Ikeda, Y.; Tsuji, A.; Matsuda, S. A new concept of associations between gut microbiota, immunity and central nervous system for the innovative treatment of neurodegenerative disorders. Metabolites 2022, 12, 1052. [Google Scholar] [CrossRef] [PubMed]
  61. Marano, G.; Mazza, M.; Lisci, F.M.; Ciliberto, M.; Traversi, G.; Kotzalidis, G.D.; De Berardis, D.; Laterza, L.; Sani, G.; Gasbarrini, A.; et al. The Microbiota-Gut-Brain axis: Psychoneuroimmunological insights. Nutrients 2023, 15, 1496. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, S.; Luo, S.; Yan, C. Gut microbiota implications for health and welfare in farm animals, A review. Animals 2021, 12, 93. [Google Scholar] [CrossRef] [PubMed]
  63. Cheng, L.; Wu, H.; Chen, Z.; Hao, H.; Zheng, X. Gut microbiome at the crossroad of genetic variants and behavior disorders. Gut Microbes 2023, 15, 2201156. [Google Scholar] [CrossRef] [PubMed]
  64. Chowdhury, M.A.H.; Ashrafudoulla, M.; Mevo, S.I.U.; Mizan, M.F.R.; Park, S.H.; Ha, S.D. Current and future interventions for improving poultry health and poultry food safety and security: A comprehensive review. Compr. Rev. Food Sci. Food Saf. 2023, 22, 1555–1596. [Google Scholar] [CrossRef]
  65. Chen, P.; Zhang, L.; Feng, Y.; Liu, Y.-F.; Si, T.L.; Su, Z.; Cheung, T.; Ungvari, G.S.; Ng, C.H.; Xiang, Y.-T. Brain-gut axis and psychiatric disorders: A perspective from bibliometric and visual analysis. Front. Immunol. 2022, 13, 1047007. [Google Scholar] [CrossRef] [PubMed]
  66. Demin, K.A.; Zabegalov, K.A.; Kolesnikova, T.O.; Galstyan, D.S.; Kositsyn, Y.M.; Costa, F.V.; De Abreu, M.S.; Kalueff, A.V. Animal Inflammation-Based Models of Neuropsychiatric Disorders. In Neuroinflammation, Gut-Brain Axis and Immunity in Neuropsychiatric Disorders; Springer Nature: Singapore, 2023; pp. 91–104. [Google Scholar]
  67. Mitrea, L.; Nemeş, S.A.; Szabo, K.; Teleky, B.E.; Vodnar, D.C. Guts Imbalance Imbalances the Brain: A Review of Gut Microbiota Association with Neurological and Psychiatric Disorders. Front. Med. 2022, 9, 813204. [Google Scholar] [CrossRef] [PubMed]
  68. Carloni, S.; Rescigno, M. The gut-brain vascular axis in neuroinflammation. Semin. Immunol. 2023, 69, 101802. [Google Scholar] [CrossRef]
  69. Cheng, J.; Lei, H.; Xie, C.; Chen, J.; Yi, X.; Zhao, F.; Yuan, Y.; Chen, P.; He, J.; Luo, C.; et al. B lymphocyte development in the bursa of fabricius of young broilers is influenced by the gut microbiota. Microbiol. Spectr. 2023, 11, e0479922. [Google Scholar] [CrossRef]
  70. Huang, C.; Hao, E.; Yue, Q.; Liu, M.; Wang, D.; Chen, Y.; Shi, L.; Zeng, D.; Zhao, G.; Chen, H. Malfunctioned inflammatory response and serotonin metabolism at the microbiota-gut-brain axis drive feather pecking behavior in laying hens. Poult. Sci. 2023, 102, 102686. [Google Scholar] [CrossRef]
  71. Li, Y.; Yang, L.; Li, J.; Gao, W.; Zhao, Z.; Dong, K.; Duan, W.; Dai, B.; Guo, R. Antidepression of Xingpijieyu formula targets gut microbiota derived from depressive disorder. CNS Neurosci. Ther. 2023, 29, 669–681. [Google Scholar] [CrossRef]
  72. Kim, J.E.; Tun, H.M.; Bennett, D.C.; Leung, F.C.; Cheng, K.M. Microbial diversity and metabolic function in duodenum, jejunum and ileum of emu (Dromaius novaehollandiae). Sci. Rep. 2023, 13, 4488. [Google Scholar] [CrossRef]
  73. Koyasu, H.; Takahashi, H.; Yoneda, M.; Naba, S.; Sakawa, N.; Sasao, I.; Nagasawa, M.; Kikusui, T. Correlations between behavior and hormone concentrations or gut microbiome imply that domestic cats (Felis silvestris catus) living in a group are not like ‘groupmates’. PLoS ONE 2022, 17, e0269589. [Google Scholar] [CrossRef] [PubMed]
  74. Queiroz, S.A.L.; Ton, A.M.M.; Pereira, T.M.C.; Campagnaro, B.P.; Martinelli, L.; Picos, A.; Campos-Toimil, M.; Vasquez, E.C. The gut microbiota-brain axis: A new frontier on neuropsychiatric disorders. Front. Psychiatry 2022, 13, 872594. [Google Scholar] [CrossRef] [PubMed]
  75. Accettulli, A.; Corbo, M.R.; Sinigaglia, M.; Speranza, B.; Campaniello, D.; Racioppo, A.; Altieri, C.; Bevilacqua, A. Psycho-microbiology, a new frontier for probiotics: An exploratory overview. Microorganisms 2022, 10, 2141. [Google Scholar] [CrossRef] [PubMed]
  76. Bhatia, N.Y.; Jalgaonkar, M.P.; Hargude, A.B.; Sherje, A.P.; Oza, M.J.; Doshi, G.M. Gut-Brain axis and neurological disorders-how microbiomes affect our mental health. CNS Neurol. Disord. -Drug Targets 2023, 22, 1008–1030. [Google Scholar] [CrossRef] [PubMed]
  77. Garvey, M. The association between dysbiosis and neurological conditions often manifesting with chronic pain. Biomedicines 2023, 11, 748. [Google Scholar] [CrossRef]
  78. Handajani, Y.S.; Hengky, A.; Schröder-Butterfill, E.; Hogervorst, E.; Turana, Y. Probiotic supplementation improved cognitive function in cognitively impaired and healthy older adults: A systematic review of recent trials. Neurol. Sci. 2023, 44, 1163–1169. [Google Scholar] [CrossRef]
  79. Johnson, D.; Letchumanan, V.; Thum, C.C.; Thurairajasingam, S.; Lee, L.H. A microbial-based approach to mental health: The potential of probiotics in the treatment of depression. Nutrients 2023, 15, 1382. [Google Scholar] [CrossRef]
  80. Kim, I.B.; Park, S.C.; Kim, Y.K. Microbiota-Gut-Brain axis in major depression: A new therapeutic approach. Adv. Exp. Med. Biol. 2023, 1411, 209–224. [Google Scholar]
  81. Mohan, A.; Godugu, S.; Joshi, S.S.; Shah, K.B.; Vanka, S.C.; Shakil, H.; Dhanush, P.; Veliginti, S.; Sure, P.S.; Goranti, J. Gut-brain axis: Altered microbiome and depression—Review. Ann. Med. Surg. 2023, 85, 1784–1789. [Google Scholar]
  82. Rathour, D.; Shah, S.; Khan, S.; Singh, P.K.; Srivastava, S.; Singh, S.B.; Khatri, D.K. Role of gut microbiota in depression: Understanding molecular pathways, recent research, and future direction. Behav. Brain Res. 2023, 436, 114081. [Google Scholar] [CrossRef] [PubMed]
  83. Sikorska, M.; Antosik-Wójcińska, A.Z.; Dominiak, M. Probiotics as a tool for regulating molecular mechanisms in depression: A systematic review and meta-analysis of randomized clinical trials. Int. J. Mol. Sci. 2023, 24, 3081. [Google Scholar] [CrossRef] [PubMed]
  84. Varesi, A.; Campagnoli, L.I.M.; Chirumbolo, S.; Candiano, B.; Carrara, A.; Ricevuti, G.; Esposito, C.; Pascale, A. The brain-gut-microbiota interplay in depression: A key to design innovative therapeutic approaches. Pharmacol. Res. 2023, 192, 106799. [Google Scholar] [CrossRef]
  85. Hashimoto, K. Emerging role of the host microbiome in neuropsychiatric disorders: Overview and future directions. Mol. Psychiatry. 2023, 28, 3625–3637. [Google Scholar] [CrossRef] [PubMed]
  86. Dinan, K.; Dinan, T.G. Gut microbes and neuropathology: Is there a causal nexus? Pathogens 2022, 11, 796. [Google Scholar] [CrossRef] [PubMed]
  87. Johnson, K.V.; Watson, K.K.; Dunbar, R.I.M.; Burnet, P.W.J. Sociability in a non-captive macaque population is associated with beneficial gut bacteria. Front. Microbiol. 2022, 13, 1032495. [Google Scholar] [CrossRef]
  88. Clench, M.H.; Mathias, J.R. The Avian cecum: A review. Wilson Bull. 1995, 107, 93–121. [Google Scholar]
  89. Stanley, D.; Geier, M.S.; Chen, H.; Hughes, R.J.; Moore, R.J. Comparison of fecal and cecal microbiotas reveals qualitative similarities but quantitative differences. BMC Microbiol. 2015, 15, 51. [Google Scholar] [CrossRef]
  90. Hunt, A.; Al-Nakkash, L.; Lee, A.H.; Smith, H.F. Phylogeny and herbivory are related to avian cecal size. Sci. Rep. 2019, 9, 4243. [Google Scholar] [CrossRef]
  91. Di Marcantonio, L.; Marotta, F.; Vulpiani, M.P.; Sonntag, Q.; Iannetti, L.; Janowicz, A.; Di Serafino, G.; Di Giannatale, E.; Garofolo, G. Investigating the cecal microbiota in broiler poultry farms and its potential relationships with animal welfare. Res. Vet. Sci. 2022, 144, 115–125. [Google Scholar] [CrossRef] [PubMed]
  92. Campos, P.M.; Schreier, L.L.; Proszkowiec-Weglarz, M.; Dridi, S. Cecal microbiota composition differs under normal and high ambient temperatures in genetically distinct chicken lines. Sci. Rep. 2023, 13, 16037. [Google Scholar] [CrossRef]
  93. Cazals, A.; Estellé, J.; Bruneau, N.; Coville, J.L.; Menanteau, P.; Rossignol, M.N.; Jardet, D.; Bevilacqua, C.; Rau, A.; Bed’Hom, B.; et al. Differences in caecal microbiota composition and Salmonella carriage between experimentally infected inbred lines of chickens. Genet. Sel. Evol. 2022, 54, 7. [Google Scholar] [CrossRef]
  94. Van der Eijk, J.A.; Lammers, A.; Kjaer, J.B.; Rodenburg, T.B. Stress response, peripheral serotonin and natural antibodies in feather pecking genotypes and phenotypes and their relation with coping style. Physiol. Behav. 2019, 199, 1–10. [Google Scholar] [CrossRef]
  95. Van der Eijk, J.A.; Verwoolde, M.B.; de Vries Reilingh, G.; Jansen, C.A.; Rodenburg, T.B.; Lammers, A. Chicken lines divergently selected on feather pecking differ in immune characteristics. Physiol. Behav. 2019, 212, 112680. [Google Scholar] [CrossRef]
  96. Meyer, B.; Zentek, J.; Harlander-Matauschek, A. Differences in intestinal microbial metabolites in laying hens with high and low levels of repetitive feather-pecking behavior. Physiol. Behav. 2013, 110, 96–101. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, C.; Li, Y.; Wang, H.; Li, M.; Rong, J.; Liao, X.; Wu, Y.; Wang, Y. Differences in peripheral and central metabolites and gut microbiome of laying hens with different feather-pecking phenotypes. Front. Microbiol. 2023, 14, 1132866. [Google Scholar] [CrossRef]
  98. Borda-Molina, D.; Iffland, H.; Schmid, M.; Müller, R.; Schad, S.; Seifert, J.; Tetens, J.; Bessei, W.; Bennewitz, J.; Camarinha-Silva, A. Gut microbial composition and predicted functions are not associated with feather pecking and antagonistic behavior in laying hens. Life 2021, 11, 235. [Google Scholar] [CrossRef] [PubMed]
  99. Rubio, L.A. Possibilities of early life programming in broiler chickens via intestinal microbiota modulation. Poult. Sci. 2019, 98, 695–706. [Google Scholar] [CrossRef]
  100. Bacon, L.D.; Hunt, H.D.; Cheng, H.H. Genetic resistance to Marek’s disease. Curr. Top. Microbiol. Immunol. 2001, 255, 121–141. [Google Scholar]
  101. Boodhoo, N.; Gurung, A.; Sharif, S.; Behboudi, S. Marek’s disease in chickens: A review with focus on immunology. Vet. Res. 2016, 47, 119. [Google Scholar] [CrossRef] [PubMed]
  102. Xu, L.; He, Y.; Ding, Y.; Liu, G.E.; Zhang, H.; Cheng, H.H.; Taylor, R.L.; Song, J. Genetic assessment of inbred chicken lines indicates genomic signatures of resistance to Marek’s disease. J. Anim. Sci. Biotechnol. 2018, 9, 1–10. [Google Scholar] [CrossRef] [PubMed]
  103. Dennis, R.L.; Zhang, H.W.; Bacon, L.D.; Estevez, I.; Cheng, H.W. Behavioral and physiological features of chickens diversely selected for resistance to avian disease: I. Selected inbred lines differ for behavioral and physical responses to social stress. Poult. Sci. 2004, 83, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
  104. Dennis, R.L.; Zhang, H.M.; Cheng, H.W. Effect of selection for resistance and susceptibility to viral diseases on concentrations of dopamine and immunological parameters in six-week-old chickens. Poult. Sci. 2006, 85, 2135–2140. [Google Scholar] [CrossRef]
  105. Dennis, R.L.; Cheng, H.W. Differential serotonergic mediation of aggression in roosters bred for resistance and susceptibility to Marek’s disease. Br. Poult. Sci. 2014, 55, 13–20. [Google Scholar] [CrossRef]
  106. Fu, Y.; Hu, J.Y.; Erasmus, M.; Johnson, T.; Cheng, H.W. Effects of early-life cecal microbiota transplantation from divergently selected inbred chicken lines on growth, gut serotonin, and immune parameters in recipient chickens. Poult. Sci. 2022, 101, 101925. [Google Scholar] [CrossRef]
  107. Bacon, L.D.; Hunt, H.D.; Cheng, H.H. A review of the development of chicken lines to resolve genes determining resistance to diseases. Poult. Sci. 2000, 79, 1082–1093. [Google Scholar] [CrossRef] [PubMed]
  108. Bacon, L.D.; Palmquist, D. Chicken lines differ in production of interferon-like activity by peripheral white blood cells stimulated with phytohemagglutinin. Poult. Sci. 2002, 81, 1629–1636. [Google Scholar] [CrossRef]
  109. Yonash, N.I.; Bacon, L.D.; Smith, E.J. Concentration of immunoglobulin G in plasma varies among 6C. 7 recombinant congenic strains of chickens. Poult. Sci. 2002, 81, 1104–1108. [Google Scholar] [CrossRef]
  110. Warren, W.C.; Rice, E.S.; Meyer, A.; Hearn, C.J.; Steep, A.; Hunt, H.D.; Monson, M.S.; Lamont, S.J.; Cheng, H.H. The immune cell landscape and response of Marek’s disease resistant and susceptible chickens infected with Marek’s disease virus. Sci. Rep. 2023, 13, 5355. [Google Scholar] [CrossRef]
  111. Dennis, R.L.; Muir, M.W.; Cheng, H.W. Effects of raclopride on aggression and stress in diversely selected chicken lines. Behav. Brain Res. 2006, 175, 104–111. [Google Scholar] [CrossRef]
  112. Hu, J.Y.; Johnson, T.A.; Zhang, H.; Cheng, H.W. The microbiota-gut-brain axis: Gut microbiota modulates conspecific aggression in diversely selected laying hens. Microorganisms 2022, 10, 1081. [Google Scholar] [CrossRef]
  113. Burgess, S.C.; Basaran, B.H.; Davison, T.F. Resistance to marek’s disease herpesvirus-induced lymphoma is multiphasic and dependent on host genotype. Vet. Pathol. 2002, 38, 129–142. [Google Scholar] [CrossRef]
  114. Perumbakkam, S.; Hunt, H.D.; Cheng, H.H. Differences in CD8αα and cecal microbiome community during proliferation and late cytolytic phases of Marek’s disease virus infection are associated with genetic resistance to Marek’s disease. FEMS Microbiol. Ecol. 2016, 92, 188. [Google Scholar] [CrossRef] [PubMed]
  115. Heidari, M.; Wang, D.; Delekta, P.; Sun, S. Marek’s disease virus immunosuppression alters host cellular responses and immune gene expression in the skin of infected chickens. Vet. Immunol. Immunopathol. 2016, 180, 21–28. [Google Scholar] [CrossRef] [PubMed]
  116. Kaiser, P.; Underwood, G.; Davison, F. Differential cytokine responses following Marek’s disease virus infection of chickens differing in resistance to Marek’s disease. J. Virol. 2003, 77, 762–768. [Google Scholar] [CrossRef] [PubMed]
  117. Haunshi, S.; Cheng, H.H. Differential expression of Toll-like receptor pathway genes in chicken embryo fibroblasts from chickens resistant and susceptible to Marek’s disease. Poult. Sci. 2014, 93, 550–555. [Google Scholar] [CrossRef] [PubMed]
  118. De Haas, E.N.; van der Eijk, J.A. Where in the serotonergic system does it go wrong? Unravelling the route by which the serotonergic system affects feather pecking in chickens. Neurosci. Biobehav. Rev. 2018, 95, 170–188. [Google Scholar] [CrossRef] [PubMed]
  119. Jadhav, V.V.; Han, J.; Fasina, Y.; Harrison, S.H. Connecting gut microbiomes and short chain fatty acids with the serotonergic system and behavior in Gallus gallus and other avian species. Front. Physiol. 2022, 13, 1035538. [Google Scholar] [CrossRef]
  120. Krakowski, M. Violence and Serotonin: Influence of Impulse Control, Affect Regulation, and Social Functioning. J. Neuropsych. Clin. Neurosci. 2003, 15, 3. [Google Scholar] [CrossRef] [PubMed]
  121. Blake, P.; Grafman, J. The neurobiology of aggression. Lancet 2004, 364, 12–13. [Google Scholar] [CrossRef]
  122. Da Cunha-Bang, S.; Knudsen, G.M. The Modulatory Role of Serotonin on Human Impulsive Aggression. Biol. Psychiatry 2021, 90, 447–457. [Google Scholar] [CrossRef]
  123. Fu, Y.; Hu, J.; Erasmus, M.A.; Zhang, H.; Johnson, T.A.; Cheng, H. Cecal microbiota transplantation: Unique influence of cecal microbiota from two divergently selected inbred donor lines on cecal microbial profile, serotonergic activity, and aggressive behavior of recipient chickens. J. Anim. Sci. Biotechnol. 2023, 14, 1–16. [Google Scholar]
  124. Webster, A.B. Behavior of chickens. In Commercial Chicken Meat and Egg Production; Bell, D.D., Weaver, W.D., Eds.; Springer: Boston, MA, USA, 2002. [Google Scholar]
  125. Dennis, R.L.; Fahey, A.G.; Cheng, H.W. Alterations to embryonic serotonin change aggression and fearfulness. Aggress. Behav. 2013, 39, 91–98. [Google Scholar] [CrossRef]
  126. Daigle, C.L.; Rodenburg, T.B.; Bolhuis, J.E.; Swanson, J.C.; Siegford, J.M. Use of dynamic and rewarding environmental enrichment to alleviate feather pecking in non-cage laying hens. Appl. Anim. Behav. Sci. 2014, 161, 75–85. [Google Scholar] [CrossRef]
  127. Dennis, R.L.; Cheng, H.W. The dopaminergic system and aggression in laying hens. Poult. Sci. 2011, 90, 2440–2448. [Google Scholar] [CrossRef]
  128. Dennis, R.L.; Lay, D.J., Jr.; Cheng, H.W. Effects of early serotonin programming on behavioral and central monoamine concentrations in an avian model. Behav. Brain Res. 2013, 253, 290–296. [Google Scholar] [CrossRef] [PubMed]
  129. Lourenço-Silva, M.I.; Ulans, A.; Campbell, A.M.; Almeida, I.C.L.; Jacobs, L. Social-pair judgment bias testing in slow-growing broiler chickens raised in low- or high-complexity environments. Sci. Rep. 2023, 13, 9393. [Google Scholar] [CrossRef] [PubMed]
  130. Koolhaas, J.M.; Coppens, C.M.; de Boer, S.F.; Buwalda, B.; Meerlo, P.; Timmermans, P.J. The resident-intruder paradigm: A standardized test for aggression, violence and social stress. J. Vis. Exp. 2013, 77, e4367. [Google Scholar]
  131. Foster, J.A.; Rinaman, L.; Cryan, J.F. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol. Stress 2017, 7, 124–136. [Google Scholar] [PubMed]
  132. Freimer, D.; Yang, T.T.; Ho, T.C.; Tymofiyeva, O.; Leung, C. The gut microbiota, HPA axis, and brain in adolescent-onset depression: Probiotics as a novel treatment. Brain Behav. Immun. Health 2022, 26, 100541. [Google Scholar] [CrossRef] [PubMed]
  133. Senchukova, M.A. Microbiota of the gastrointestinal tract: Friend or foe? World J. Gastroenterol. 2023, 29, 19–42. [Google Scholar] [CrossRef] [PubMed]
  134. Shakhar, K. The inclusive behavioral immune system. Front. Psychol. 2019, 10, 1004. [Google Scholar] [CrossRef]
  135. Parashar, A.; Udayabanu, M. Gut microbiota regulates key modulators of social behavior. Eur. Neuropsychopharmacol. 2016, 26, 78–91. [Google Scholar] [CrossRef]
  136. Stilling, R.M.; Bordenstein, S.R.; Dinan, T.G.; Cryan, J.F. Friends with social benefits: Host-microbe interactions as a driver of brain evolution and development? Front. Cell Infect. Microbiol. 2014, 4, 147. [Google Scholar] [CrossRef]
  137. Gulledge, L.; Oyebode, D.; Donaldson, J.R. The influence of the microbiome on aggressive behavior: An insight into age-related aggression. FEMS Microbiol. Lett. 2023, 370, fnac114. [Google Scholar] [CrossRef]
  138. Johnson, K.V. Gut microbiome composition and diversity are related to human personality traits. Hum. Microb. J. 2020, 15, 100069. [Google Scholar] [CrossRef]
  139. Aatsinki, A.K.; Lahti, L.; Uusitupa, H.M.; Munukka, E.; Keskitalo, A.; Nolvi, S.; O’Mahony, S.; Pietilä, S.; Elo, L.L.; Eerola, E.; et al. Gut microbiota composition is associated with temperament traits in infants. Brain Behav. Immun. 2019, 80, 849–858. [Google Scholar] [CrossRef] [PubMed]
  140. Alving-Jessep, E.; Botchway, E.; Wood, A.G.; Hilton, A.C.; Blissett, J.M. The development of the gut microbiome and temperament during infancy and early childhood: A systematic review. Dev. Psychobiol. 2022, 64, e22306. [Google Scholar] [CrossRef]
  141. Saleena, L.A.K.; Teo, M.Y.M.; How, Y.H.; In, L.L.A.; Pui, L.P. Immunomodulatory action of Lactococcus lactis. J. Biosci. Bioeng. 2023, 135, 1–9. [Google Scholar] [CrossRef]
  142. Obianwuna, U.E.; Agbai Kalu, N.; Wang, J.; Zhang, H.; Qi, G.; Qiu, K.; Wu, S. Recent trends on mitigative effect of probiotics on oxidative-stress-induced gut dysfunction in broilers under necrotic enteritis challenge: A review. Antioxidants 2023, 12, 911. [Google Scholar] [CrossRef] [PubMed]
  143. Vaccaro, R.; Casini, A.; Severi, C.; Lamazza, A.; Pronio, A.; Palma, R. Serotonin and melatonin in human lower gastrointestinal tract. Diagnostics 2023, 13, 204. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, H.; Wang, Z.; Wang, G.; Song, X.; Qian, Y.; Liao, Z.; Sui, L.; Ai, L.; Xia, Y. Understanding the connection between gut homeostasis and psychological stress. J. Nutr. 2023, 153, 924–939. [Google Scholar] [CrossRef] [PubMed]
  145. Soares, I.; Belote, B.L.; Santin, E.; Dal Pont, G.C.; Kogut, M.H. Morphological assessment and biomarkers of low-grade, chronic intestinal inflammation in production animals. Animals 2022, 12, 3036. [Google Scholar] [CrossRef]
  146. Riehl, L.; Furst, J.; Kress, M.; Rykalo, N. The importance of the gut microbiome and its signals for a healthy nervous system and the multifaceted mechanisms of neuropsychiatric disorders. Front. Neurosci. 2023, 17, 1302957. [Google Scholar] [CrossRef] [PubMed]
  147. Boehme, M.; Guzzetta, K.E.; Wasén, C.; Cox, L.M. The gut microbiota is an emerging target for improving brain health during ageing. Gut Microbiome 2023, 4, E2. [Google Scholar] [CrossRef]
  148. Laman, J.D. Cutting edge technologies in chronic inflammation research. Exp. Dermatol. 2022, 31, 17–21. [Google Scholar] [CrossRef]
  149. Wu, W.L.; Adame, M.D.; Liou, C.W.; Barlow, J.T.; Lai, T.T.; Sharon, G.; Schretter, C.E.; Needham, B.D.; Wang, M.I.; Tang, W.; et al. Microbiota regulate social behaviour via stress response neurons in the brain. Nature 2021, 595, 409–414. [Google Scholar] [CrossRef]
  150. Kasarello, K.; Cudnoch-Jedrzejewska, A.; Czarzasta, K. Communication of gut microbiota and brain via immune and neuroendocrine signaling. Front. Microbiol. 2023, 14, 1118529. [Google Scholar] [CrossRef]
  151. Bhatt, S.; Kanoujia, J.; Mohana Lakshmi, S.; Patil, C.R.; Gupta, G.; Chellappan, D.K.; Dua, K. Role of brain-gut-microbiota axis in depression: Emerging therapeutic avenues. CNS Neurol. Disord. -Drug Targets 2023, 22, 276–288. [Google Scholar] [CrossRef]
  152. Dinan, T.G.; Cryan, J.F. Regulation of the stress response by the gut microbiota: Implications for psychoneuroendocrinology. Psychoneuroendocrinology 2012, 37, 1369–1378. [Google Scholar] [CrossRef] [PubMed]
  153. Ilchmann-Diounou, H.; Menard, S. Psychological stress, intestinal barrier dysfunctions, and autoimmune disorders: An overview. Front. Immunol. 2020, 11, 1823. [Google Scholar] [CrossRef]
  154. Feng, S.; Meng, C.; Liu, Y.; Yi, Y.; Liang, A.; Zhang, Y.; Hao, Z. Bacillus licheniformis prevents and reduces anxiety-like and depression-like behaviours. Appl. Microbiol. Biotechnol. 2023, 107, 4355–4368. [Google Scholar] [CrossRef] [PubMed]
  155. Bloemendaal, M.; Szopinska-Tokov, J.; Belzer, C.; Boverhoff, D.; Papalini, S.; Michels, F.; van Hemert, S.; Arias Vasquez, A.; Aarts, E. Probiotics-induced changes in gut microbial composition and its effects on cognitive performance after stress: Exploratory analyses. Transl. Psychiatry 2021, 11, 300. [Google Scholar] [CrossRef] [PubMed]
  156. De Haas, E.N.; Newberry, R.C.; Edgar, J.; Riber, A.B.; Estevez, I.; Ferrante, V.; Hernandez, C.E.; Kjaer, J.B.; Ozkan, S.; Dimitrov, I.; et al. Prenatal and early postnatal behavioural programming in laying hens, with possible implications for the development of injurious pecking. Front. Vet. Sci. 2021, 8, 678500. [Google Scholar] [CrossRef] [PubMed]
  157. Kraimi, N.; Dawkins, M.; Gebhardt-Henrich, S.G.; Velge, P.; Rychlik, I.; Volf, J.; Creach, P.; Smith, A.; Colles, F.; Leterrier, C. Influence of the microbiota-gut-brain axis on behavior and welfare in farm animals: A review. Physiol. Behav. 2019, 210, 112658. [Google Scholar] [CrossRef] [PubMed]
  158. Davidson, G.L.; Raulo, A.; Knowles, S.C. Identifying Microbiome-Mediated Behaviour in Wild Vertebrates. Trends Ecol. Evol. 2020, 35, 972–980. [Google Scholar] [CrossRef]
  159. Mikami, K.; Watanabe, N.; Tochio, T.; Kimoto, K.; Akama, F.; Yamamoto, K. Impact of Gut Microbiota on Host Aggression: Potential Applications for Therapeutic Interventions Early in Development. Microorganisms 2023, 11, 1008. [Google Scholar] [CrossRef]
  160. Lalonde, R.; Strazielle, C. Probiotic effects on anxiety-like behavior in animal models. Rev. Neurosci. 2022, 33, 691–701. [Google Scholar] [CrossRef]
  161. Huang, C.; Yue, Q.; Sun, L.; Di, K.; Yang, D.; Hao, E.; Wang, D.; Chen, Y.; Shi, L.; Zhou, R.; et al. Restorative effects of Lactobacillus rhamnosus LR-32 on the gut microbiota, barrier integrity, and 5-HT metabolism in reducing feather-pecking behavior in laying hens with antibiotic-induced dysbiosis. Front. Microbiol. 2023, 14, 1173804. [Google Scholar] [CrossRef]
  162. Villageliu, D.N.; Lyte, M. Microbial endocrinology: Why the intersection of microbiology and neurobiology matters to poultry health. Poult. Sci. 2017, 96, 2501–2508. [Google Scholar] [CrossRef]
  163. Sun, W.; Zhang, L. Antioxidant indexes and immune function of the intestinal flora of compound microecological preparations. Oxidative Med. Cell. Longev. 2022, 2022, 5498514. [Google Scholar] [CrossRef]
  164. Birkl, P.; Bharwani, A.; Kjaer, J.B.; Kunze, W.; McBride, P.; Forsythe, P.; Harlander-Matauschek, A. Differences in cecal microbiome of selected high and low feather-pecking laying hens. Poult. Sci. 2018, 97, 3009–3014. [Google Scholar] [CrossRef]
  165. Campbell, C.; Kandalgaonkar, M.R.; Golonka, R.M.; Yeoh, B.S.; Vijay-Kumar, M.; Saha, P. Crosstalk between gut microbiota and host immunity: Impact on inflammation and immunotherapy. Biomedicines 2023, 11, 294. [Google Scholar] [CrossRef] [PubMed]
  166. Jach, M.E.; Serefko, A.; Szopa, A.; Sajnaga, E.; Golczyk, H.; Santos, L.S.; Borowicz-Reutt, K.; Sieniawska, E. The role of probiotics and their metabolites in the treatment of depression. Molecules 2023, 28, 3213. [Google Scholar] [CrossRef] [PubMed]
  167. Mazziotta, C.; Tognon, M.; Martini, F.; Torreggiani, E.; Rotondo, J.C. Probiotics mechanism of action on immune cells and beneficial effects on human health. Cells 2023, 12, 184. [Google Scholar] [CrossRef] [PubMed]
  168. Ribaldone, D.G.; Pellicano, R.; Fagoonee, S.; Actis, G.C. Modulation of the gut microbiota: Opportunities and regulatory aspects. Minerva Gastroenterol. 2023, 69, 128–140. [Google Scholar] [CrossRef] [PubMed]
  169. Zhang, Y.; Zhang, Y.; Liu, F.; Mao, Y.; Zhang, Y.; Zeng, H.; Ren, S.; Guo, L.; Chen, Z.; Hrabchenko, N.; et al. Mechanisms and applications of probiotics in prevention and treatment of swine diseases. Porcine Health Manag. 2023, 9, 5. [Google Scholar] [CrossRef] [PubMed]
  170. Zheng, Y.; Zhang, Z.; Tang, P.; Wu, Y.; Zhang, A.; Li, D.; Wang, C.Z.; Wan, J.Y.; Yao, H.; Yuan, C.S. Probiotics fortify intestinal barrier function: A systematic review and meta-analysis of randomized trials. Front. Immunol. 2023, 14, 1143548. [Google Scholar] [CrossRef] [PubMed]
  171. Collins, S.M.; Kassam, Z.; Bercik, P. The adoptive transfer of behavioral phenotype via the intestinal microbiota: Experimental evidence and clinical implications. Curr. Opin. Microbiol. 2013, 16, 240–245. [Google Scholar] [CrossRef] [PubMed]
  172. Ait-Belgnaoui, A.; Colom, A.; Braniste, V.; Ramalho, L.; Marrot, A.; Cartier, C.; Houdeau, E.; Theodorou, V.; Tompkins, T. Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterol. Motil. 2014, 26, 510–520. [Google Scholar] [CrossRef]
  173. Parois, S.; Calandreau, L.; Kraimi, N.; Gabriel, I.; Leterrier, C. The influence of a probiotic supplementation on memory in quail suggests a role of gut microbiota on cognitive abilities in birds. Behav. Brain Res. 2017, 331, 47–53. [Google Scholar] [CrossRef]
  174. Naglaa, M. Do probiotics affect the behavior of turkey poults? J. Vet. Med. Anim. Health 2013, 5, 144–148. [Google Scholar]
  175. Mindus, C.; van Staaveren, N.; Bharwani, A.; Fuchs, D.; Gostner, J.M.; Kjaer, J.B.; Kunze, W.; Mian, M.F.; Shoveller, A.K.; Forsythe, P.; et al. Ingestion of Lactobacillus rhamnosus modulates chronic stress-induced feather pecking in chickens. Sci. Rep. 2021, 11, 17119. [Google Scholar] [CrossRef] [PubMed]
  176. Mindus, C.; van Staaveren, N.; Fuchs, D.; Gostner, J.M.; Kjaer, J.B.; Kunze, W.; Mian, M.F.; Shoveller, A.K.; Forsythe, P.; Harlander-Matauschek, A. Regulatory T cell modulation by lactobacillus rhamnosus improves feather damage in chickens. Front. Vet. Sci. 2022, 9, 855261. [Google Scholar] [CrossRef] [PubMed]
  177. Jiang, S.; Hu, J.Y.; Cheng, H.W. The Impact of Probiotic Bacillus subtilis on Injurious Behavior in Laying Hens. Animals 2022, 12, 870. [Google Scholar] [CrossRef] [PubMed]
  178. Allegretti, J.R.; Kelly, C.R.; Grinspan, A.; Mullish, B.H.; Hurtado, J.; Carrellas, M.; Marcus, J.; Marchesi, J.R.; McDonald, J.A.; Gerardin, Y.; et al. Inflammatory Bowel Disease Outcomes Following Fecal Microbiota Transplantation for Recurrent C. difficile Infection. Inflamm. Bowel Dis. 2021, 27, 1371–1378. [Google Scholar] [CrossRef] [PubMed]
  179. Tariq, R.; Syed, T.; Yadav, D.; Prokop, L.J.; Singh, S.; Loftus, E.V., Jr.; Pardi, D.S.; Khanna, S. Outcomes of fecal microbiota transplantation for C. difficile infection in inflammatory bowel disease: A systematic review and meta-analysis. J. Clin. Gastroenterol. 2023, 57, 285–293. [Google Scholar] [CrossRef]
  180. Anand, N.; Gorantla, V.R.; Chidambaram, S.B. The Role of Gut Dysbiosis in the Pathophysiology of Neuropsychiatric Disorders. Cells 2022, 12, 54. [Google Scholar] [CrossRef]
  181. Hamamah, S.; Gheorghita, R.; Lobiuc, A.; Sirbu, I.O.; Covasa, M. Fecal microbiota transplantation in non-communicable diseases: Recent advances and protocols. Front. Med. 2022, 9, 1060581. [Google Scholar] [CrossRef]
  182. Qi, R.; Zhang, Z.; Wang, J.; Qiu, X.; Wang, Q.; Yang, F.; Huang, J.; Liu, Z. Introduction of colonic and fecal microbiota from an adult pig differently affects the growth, gut health, intestinal microbiota and blood metabolome of newborn piglets. Front. Microbiol. 2021, 12, 623673. [Google Scholar] [CrossRef] [PubMed]
  183. Shehata, E.; Parker, A.; Suzuki, T.; Swann, J.R.; Suez, J.; Kroon, P.A.; Day-Walsh, P. Microbiomes in physiology: Insights into 21st-century global medical challenges. Exp. Physiol. 2022, 107, 257–264. [Google Scholar] [CrossRef] [PubMed]
  184. Medel-Matus, J.S.; Shin, D.; Dorfman, E.; Sankar, R.; Mazarati, A. Facilitation of kindling epileptogenesis by chronic stress may be mediated by intestinal microbiome. Epilepsia Open 2018, 3, 290–294. [Google Scholar] [CrossRef] [PubMed]
  185. Pittman, Q.J. Stress co-opts the gut to affect epileptogenesis. Commentary on “Facilitation of kindling epileptogenesis by chronic stress may be mediated by intestinal microbiome”. Epilepsia Open 2019, 4, 230–231. [Google Scholar] [CrossRef]
  186. Kragsnaes, M.S.; Kjeldsen, J.; Horn, H.C.; Munk, H.L.; Pedersen, J.K.; Just, S.A.; Ahlquist, P.; Pedersen, F.M.; de Wit, M.; Möller, S.; et al. Safety and efficacy of faecal microbiota transplantation for active peripheral psoriatic arthritis: An exploratory randomised placebo-controlled trial. Ann. Rheum. Dis. 2021, 80, 1158–1167. [Google Scholar] [CrossRef]
  187. Ting, N.L.; Lau, H.C.; Yu, J. Cancer pharmacomicrobiomics: Targeting microbiota to optimise cancer therapy outcomes. Gut 2022, 71, 1412–1425. [Google Scholar] [CrossRef]
  188. Drugs.com. Rebyota. 2022. Available online: https://www.drugs.com/rebyota.html (accessed on 30 November 2023).
  189. Pauwels, J.; Taminiau, B.; Janssens, G.P.; De Beenhouwer, M.; Delhalle, L.; Daube, G.; Coopman, F. Cecal drop reflects the chickens’ cecal microbiome, fecal drop does not. J. Microbiol. Methods 2015, 117, 164–170. [Google Scholar] [CrossRef] [PubMed]
  190. Panasevich, M.R.; Wankhade, U.D.; Chintapalli, S.V.; Shankar, K.; Rector, R.S. Cecal versus fecal microbiota in Ossabaw swine and implications for obesity. Physiol. Genom. 2018, 50, 355–368. [Google Scholar] [CrossRef]
  191. Kozik, A.J.; Nakatsu, C.H.; Chun, H.; Jones-Hall, Y.L. Comparison of the fecal, cecal, and mucus microbiome in male and female mice after TNBS-induced colitis. PLoS ONE 2019, 14, e0225079. [Google Scholar] [CrossRef]
  192. Løtvedt, P.; Fallahshahroudi, A.; Bektic, L.; Altimiras, J.; Jensen, P. Chicken domestication changes expression of stress-related genes in brain, pituitary and adrenals. Neurobiol. Stress 2017, 7, 113–121. [Google Scholar] [CrossRef]
  193. Williams, C.L.; Garcia-Reyero, N.; Martyniuk, C.J.; Tubbs, C.W.; Bisesi, J.H., Jr. Regulation of endocrine systems by the microbiome: Perspectives from comparative animal models. Gen. Comp. Endocrinol. 2020, 292, 113437. [Google Scholar] [CrossRef]
  194. Kalia, V.C.; Shim, W.Y.; Patel, S.K.S.; Gong, C.; Lee, J.K. Recent developments in antimicrobial growth promoters in chicken health: Opportunities and challenges. Sci. Total Environ. 2022, 834, 155300. [Google Scholar] [CrossRef]
  195. Lynch, C.M.K.; Cowan, C.S.M.; Bastiaanssen, T.F.S.; Moloney, G.M.; Theune, N.; van de Wouw, M.; Florensa Zanuy, E.; Ventura-Silva, A.P.; Codagnone, M.G.; Villalobos-Manríquez, F.; et al. Critical windows of early-life microbiota disruption on behaviour, neuroimmune function, and neurodevelopment. Brain Behav. Immun. 2023, 108, 309–327. [Google Scholar] [CrossRef]
  196. Goldstein, D.L. Absorption by the cecum of wild birds: Is there interspecific variation. J. Exp. Zool. 1989, 252 (Suppl. S3), 103–110. [Google Scholar] [CrossRef]
  197. Clench, M.H. The avian cecum: Update and motility review. J. Exp. Zool. 1999, 83, 441–447. [Google Scholar] [CrossRef]
  198. Svihus, B. Function of the digestive system. J. Appl. Poul. Res. 2014, 23, 306–314. [Google Scholar] [CrossRef]
  199. Svihus, B.; Choct, M.; Classen, H. Function and nutritional roles of the avian caeca: A review. Worlds Poult. Sci. J. 2013, 69, 249–264. [Google Scholar] [CrossRef]
  200. Pan, D.; Yu, Z. Intestinal microbiome of poultry and its interaction with host and diet. Gut Microbes 2014, 5, 108–119. [Google Scholar] [CrossRef] [PubMed]
  201. Rychlik, I. Composition and function of chicken gut microbiota. Animals 2020, 10, 103. [Google Scholar] [CrossRef]
  202. Plata, G.; Baxter, N.T.; Susanti, D.; Volland-Munson, A.; Gangaiah, D.; Nagireddy, A.; Mane, S.P.; Balakuntla, J.; Hawkins, T.B.; Kumar Mahajan, A. Growth promotion and antibiotic induced metabolic shifts in the chicken gut microbiome. Commun. Biol. 2022, 5, 293. [Google Scholar] [CrossRef]
  203. Such, N.; Farkas, V.; Csitári, G.; Pál, L.; Márton, A.; Menyhárt, L.; Dublecz, K. Relative effects of dietary administration of a competitive exclusion culture and a synbiotic product, age and sampling site on intestinal microbiota maturation in broiler chickens. Vet. Sci. 2021, 8, 187. [Google Scholar] [CrossRef]
  204. Xiao, S.S.; Mi, J.D.; Mei, L.; Liang, J.; Feng, K.X.; Wu, Y.B.; Liao, X.D.; Wang, Y. Microbial diversity and community variation in the intestines of layer chickens. Animals 2021, 11, 840. [Google Scholar] [CrossRef] [PubMed]
  205. Yang, T.; Jiang, Y.; Tang, J.; Chang, G.; Zhao, W.; Hou, S.; Chen, G. Comparison of cecal microbiota and performance indices between lean-type and fatty-type pekin ducks. Front. Microbiol. 2022, 13, 820569. [Google Scholar] [CrossRef] [PubMed]
  206. Konopka, A. What is microbial community ecology? ISME J. 2009, 3, 1223–1230. [Google Scholar] [CrossRef]
  207. Shah, T.; Baloch, Z.; Shah, Z.; Cui, X.; Xia, X. The intestinal microbiota: Impacts of antibiotics therapy, colonization resistance, and diseases. Int. J. Mol. Sci. 2021, 22, 6597. [Google Scholar] [CrossRef] [PubMed]
  208. Kang, K.; Hu, Y.; Wu, S.; Shi, S. Comparative metagenomic analysis of chicken gut microbial community, function, and resistome to evaluate noninvasive and cecal sampling resources. Animals 2021, 11, 1718. [Google Scholar] [CrossRef] [PubMed]
  209. Wang, B.; Du, P.; Huang, S.; He, D.; Chen, J.; Wen, X.; Yang, J.; Xian, S.; Cheng, Z. Comparison of the caecal microbial community structure and physiological indicators of healthy and infection Eimeria tenella chickens during peak of oocyst shedding. Avian Pathol. 2023, 52, 51–61. [Google Scholar] [CrossRef]
  210. Ye, J.; Yang, H.; Hu, W.; Tang, K.; Liu, A.; Bi, S. Changed cecal microbiota involved in growth depression of broiler chickens induced by immune stress. Poult. Sci. 2023, 102, 102598. [Google Scholar] [CrossRef]
  211. Yin, Z.; Ji, S.; Yang, J.; Guo, W.; Li, Y.; Ren, Z.; Yang, X. Cecal Microbial Succession and Its Apparent Association with Nutrient Metabolism in Broiler Chickens. mSphere 2023, 8, e0061422. [Google Scholar] [CrossRef]
  212. Franco, L.; Boulianne, M.; Parent, E.; Barjesteh, N.; Costa, M.C. Colonization of the gastrointestinal tract of chicks with different bacterial microbiota profiles. Animals 2023, 13, 2633. [Google Scholar] [CrossRef]
  213. Gong, Y.; Yang, H.; Wang, X.; Xia, W.; Lv, W.; Xiao, Y.; Zou, X. Early intervention with cecal fermentation broth regulates the colonization and development of gut microbiota in broiler chickens. Front. Microbiol. 2019, 10, 1422. [Google Scholar] [CrossRef]
  214. Volf, J.; Polansky, O.; Varmuzova, K.; Gerzova, L.; Sekelova, Z.; Faldynova, M.; Babak, V.; Medvecky, M.; Smith, A.L.; Kaspers, B.; et al. Transient and prolonged response of chicken cecum mucosa to colonization with different gut microbiota. PLoS ONE 2016, 11, e0163932. [Google Scholar] [CrossRef]
  215. van Hierden, Y.M.; de Boer, S.F.; Koolhaas, J.M.; Korte, S.M. The control of feather pecking by serotonin. Behav. Neurosci. 2004, 118, 575–583. [Google Scholar] [CrossRef]
  216. Falker-Gieske, C.; Bennewitz, J.; Tetens, J. The light response in chickens divergently selected for feather pecking behavior reveals mechanistic insights towards psychiatric disorders. Mol. Biol. Rep. 2022, 49, 1649–1654. [Google Scholar] [CrossRef] [PubMed]
  217. Pasquaretta, C.; Gómez-Moracho, T.; Heeb, P.; Lihoreau, M. Exploring interactions between the gut microbiota and social behavior through nutrition. Genes 2018, 9, 534. [Google Scholar] [CrossRef] [PubMed]
  218. Maraci, Ö.; Antonatou-Papaioannou, A.; Jünemann, S.; Engel, K.; Castillo-Gutiérrez, O.; Busche, T.; Kalinowski, J.; Caspers, B.A. Timing matters: Age-dependent impacts of the social environment and host selection on the avian gut microbiota. Microbiome 2022, 10, 202. [Google Scholar] [CrossRef] [PubMed]
  219. Ramírez, G.A.; Richardson, E.; Clark, J.; Keshri, J.; Drechsler, Y.; Berrang, M.E.; Meinersmann, R.J.; Cox, N.A.; Oakley, B.B. Broiler chickens and early life programming: Microbiome transplant-induced cecal community dynamics and phenotypic effects. PLoS ONE 2020, 15, e0242108. [Google Scholar] [CrossRef]
  220. Glendinning, L.; Chintoan-Uta, C.; Stevens, M.P.; Watson, M. Effect of cecal microbiota transplantation between different broiler breeds on the chick flora in the first week of life. Poult. Sci. 2022, 101, 101624. [Google Scholar] [CrossRef]
  221. Yang, Z.; Liu, X.; Wu, Y.; Peng, J.; Wei, H. Effect of the microbiome on intestinal innate immune development in early life and the potential strategy of early intervention. Front. Immunol. 2022, 13, 936300. [Google Scholar] [CrossRef]
  222. Brett, B.E.; de Weerth, C. The microbiota-gut-brain axis: A promising avenue to foster healthy developmental outcomes. Dev. Psychobiol. 2019, 61, 772–782. [Google Scholar] [CrossRef]
  223. Forssberg, H. Microbiome programming of brain development: Implications for neurodevelopmental disorders. Dev. Med. Child. Neurol. 2019, 61, 744–749. [Google Scholar] [CrossRef] [PubMed]
  224. AABP (American Association of Bovine Practitioners). Dehorning Guidelines. 2019. Available online: https://www.aabp.org/resources/aabp_guidelines/dehorning-2019.pdf (accessed on 30 November 2023).
  225. AVMA (The American Veterinary Medical Association). Supersede Policy on Castration and Dehorning of Cattle. Resolution #4—2023. 2023. Available online: https://www.avma.org/sites/default/files/2023-03/2023W_Resolution4F.pdf (accessed on 30 November 2023).
  226. AVMA (the American Veterinary Medical Association). Teeth Clipping, Tail Docking and Permanent Identification of Piglet. 2014. Available online: https://www.avma.org/sites/default/files/resources/practices_piglets_bgnd.pdf (accessed on 30 November 2023).
  227. D’Eath, R.B.; O’Driscoll, K.; Fàbrega, E. Editorial: Holistic prevention strategies for tail biting in pigs; from farm to slaughterhouse. Front. Vet. Sci. 2023, 10, 1296461. [Google Scholar] [CrossRef] [PubMed]
  228. Bleibel, L.; Dziomba, S.; Waleron, K.F.; Kowalczyk, E.; Karbownik, M.S. Deciphering psychobiotics’ mechanism of action: Bacterial extracellular vesicles in the spotlight. Front. Microbiol. 2023, 14, 1211447. [Google Scholar] [CrossRef]
  229. Ross, K. Psychobiotics: Are they the future intervention for managing depression and anxiety? A literature review. Explore 2023, 9, 669–680. [Google Scholar] [CrossRef] [PubMed]
  230. NAMI (National Alliance on Mental Illness). Mental Health by the Numbers. 2023. Available online: https://www.nami.org/mhstats (accessed on 30 November 2023).
Figure 1. Microbiota profile between two diversely selected chicken lines 63 and 72 (n = 10). (A) Faith’s PD index, values are median ± SEM, a,b indicates significant differences (p ≤ 0.05). (B) Principal coordinate analysis (PCoA) of Bray–Curtis similarity. Each dot represents one bird (n = 10), and PCo1 and PCo2 represent the percentage of variance explained by each coordinate. (C) Cecal microbial composition profiles of the recipient chickens at phylum and genus (relative abundance >2% at phylum, >1% at genus) levels. (D) DESeq2 analysis of differentially abundant ASVs between line 63 and line 72. Estimations of log2 fold change values for each ASV were computed and each point represents an ASV that was significantly different (p ≤ 0.05) [106].
Figure 1. Microbiota profile between two diversely selected chicken lines 63 and 72 (n = 10). (A) Faith’s PD index, values are median ± SEM, a,b indicates significant differences (p ≤ 0.05). (B) Principal coordinate analysis (PCoA) of Bray–Curtis similarity. Each dot represents one bird (n = 10), and PCo1 and PCo2 represent the percentage of variance explained by each coordinate. (C) Cecal microbial composition profiles of the recipient chickens at phylum and genus (relative abundance >2% at phylum, >1% at genus) levels. (D) DESeq2 analysis of differentially abundant ASVs between line 63 and line 72. Estimations of log2 fold change values for each ASV were computed and each point represents an ASV that was significantly different (p ≤ 0.05) [106].
Microorganisms 12 00471 g001
Figure 2. Timeline of the experimental design of trial 2 and trial 3 and the proposed mechanisms underlying the transplant effects on health and behavior of recipient chickens.
Figure 2. Timeline of the experimental design of trial 2 and trial 3 and the proposed mechanisms underlying the transplant effects on health and behavior of recipient chickens.
Microorganisms 12 00471 g002
Figure 3. Effects of cecal microbiota transplantation on (A) body weight of recipient roosters; ileal morphology of recipient roosters in week 5 (B) and week 16 (C). Ileal villus height (VH), crypt depth (CD), and VH/CD ratio. Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05). Abbreviations: 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control; CD, crypt depth; VH, villus height [106].
Figure 3. Effects of cecal microbiota transplantation on (A) body weight of recipient roosters; ileal morphology of recipient roosters in week 5 (B) and week 16 (C). Ileal villus height (VH), crypt depth (CD), and VH/CD ratio. Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05). Abbreviations: 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control; CD, crypt depth; VH, villus height [106].
Microorganisms 12 00471 g003
Figure 4. Effects of cecal microbiota transplantation on ileal serotonergic activities of recipient roosters. Serotonergic activity in week 5 (A,B) and week 16 (C,D). Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (P ≤ 0.05), A,B show trend differences (0.05 < P ≤ 0.10). Abbreviations: 5-HIAA, 5-hydroxuindoleacetic acid; 5-HT, serotonin; 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control [106].
Figure 4. Effects of cecal microbiota transplantation on ileal serotonergic activities of recipient roosters. Serotonergic activity in week 5 (A,B) and week 16 (C,D). Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (P ≤ 0.05), A,B show trend differences (0.05 < P ≤ 0.10). Abbreviations: 5-HIAA, 5-hydroxuindoleacetic acid; 5-HT, serotonin; 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control [106].
Microorganisms 12 00471 g004
Figure 5. Frequency of aggressive pecking of recipient chickens in week 5 and week 16. (A) Home-cage behavior. (B) Paired test. Values are means ± SEM, n = 7. * indicates significant differences (p ≤ 0.05), and # shows trend differences (0.05 < p ≤ 0.1). 63-CMT, received cecal content solution from 63 donors; 72-CMT, received cecal content solution from 72 donors; CTRL, received saline, control [123].
Figure 5. Frequency of aggressive pecking of recipient chickens in week 5 and week 16. (A) Home-cage behavior. (B) Paired test. Values are means ± SEM, n = 7. * indicates significant differences (p ≤ 0.05), and # shows trend differences (0.05 < p ≤ 0.1). 63-CMT, received cecal content solution from 63 donors; 72-CMT, received cecal content solution from 72 donors; CTRL, received saline, control [123].
Microorganisms 12 00471 g005
Figure 6. Effects of cecal microbiota transplantation on cecal microbial profiles of recipient chickens in week 5 and week 16 (n = 7). (A) Faith’s PD index, values are median ± SEM, * indicates significant differences (p ≤ 0.05), and # shows trend differences (0.05 < p ≤ 0.1). (B) Principal coordinate analysis (PCoA) of Unweighted UniFrac of recipient chickens in week 5 and week 16. Each dot represents one bird (n = 7), and PCo1 and PCo2 represent the percentage of variance explained by each coordinate. (C) DESeq2 analysis of differentially abundant ASVs between 63-CMT group and 72-CMT group in week 5 and week 16. Estimations of log2 fold change values for each ASV were computed and each point represents an ASV that was significantly different (p ≤ 0.05). 63-CMT, received cecal content solution from 63 donors; 72-CMT, received cecal content solution from 72 donors; CTRL, received saline, control [123].
Figure 6. Effects of cecal microbiota transplantation on cecal microbial profiles of recipient chickens in week 5 and week 16 (n = 7). (A) Faith’s PD index, values are median ± SEM, * indicates significant differences (p ≤ 0.05), and # shows trend differences (0.05 < p ≤ 0.1). (B) Principal coordinate analysis (PCoA) of Unweighted UniFrac of recipient chickens in week 5 and week 16. Each dot represents one bird (n = 7), and PCo1 and PCo2 represent the percentage of variance explained by each coordinate. (C) DESeq2 analysis of differentially abundant ASVs between 63-CMT group and 72-CMT group in week 5 and week 16. Estimations of log2 fold change values for each ASV were computed and each point represents an ASV that was significantly different (p ≤ 0.05). 63-CMT, received cecal content solution from 63 donors; 72-CMT, received cecal content solution from 72 donors; CTRL, received saline, control [123].
Microorganisms 12 00471 g006
Table 1. (A) Serotonergic metabolism in the raphe nucleus; (B) peripheral serotonin, tryptophan, corticosterone, and heterophil/lymphocyte ratios; and (C) peripheral immune parameters between the two divergently selected inbred chicken lines 63 and 72.
Table 1. (A) Serotonergic metabolism in the raphe nucleus; (B) peripheral serotonin, tryptophan, corticosterone, and heterophil/lymphocyte ratios; and (C) peripheral immune parameters between the two divergently selected inbred chicken lines 63 and 72.
(A)
Treatment5-HT
(ng/g)
5-HIAA
(ng/g)
5-HT/5-HIAATRP
Line 63512.6 a151.83.2 b1183.8 a
Line 72352.7 b168.94.9 a963.2 b
SEM8.212.90.222.4
p-value0.010.620.040.08
(B)
Treatment5-HT
(ng/g)
TRP
(ng/g)
CORT
(ng/mL)
H/L ratio
Line 6361.38171.52 a8.44 b0.16 b
Line 7259.46121.42 b9.75 a0.50 a
SEM3.7915.371.510.04
p-value0.730.030.05<0.0001
(C)
TreatmentIgG
(mg/mL)
IL-6
(pg/mL)
IL-2
(pg/mL)
IL-10
(pg/mL)
TNF-α
(ng/mL)
Line 6312.028.1460.099.3736.65 A
Line 7212.927.5671.6513.1330.73 B
SEM0.731.6312.81.642.37
p-value0.540.810.540.120.09
Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05), A, B indicate trend differences (0.05 < p ≤ 0.10). Abbreviations: 5-HT, serotonin; 5-HIAA, 5-hydroxuindoleacetic acid; CORT, corticosterone; H/L, heterophil-to-lymphocyte ratio; IgG, immunoglobulin G; IL, interleukin; TNF, tumor necrosis factor; TRP, tryptophan [112].
Table 2. Effects of cecal microbiota transplantation on body weight, relative organ weight, stress parameters (H/L ratio, corticosterone), and sexual hormone (testosterone) of recipient roosters in week 16.
Table 2. Effects of cecal microbiota transplantation on body weight, relative organ weight, stress parameters (H/L ratio, corticosterone), and sexual hormone (testosterone) of recipient roosters in week 16.
Measures Treatment SEMp-Value
CTRL72-CMT63-CMT
Body weight1642.51738.61711.334.30.426
Adrenal gland 14.181 AB4.762 A3.306 B0.4200.090
H/L ratio0.327 ab0.367 a0.243 b0.0290.024
Corticosterone (ng/mL)4.2354.6783.6970.9000.789
Testosterone (ng/mL)1.4231.1321.7440.2770.345
Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05), A,B indicate trend differences (0.05 < p ≤ 0.10). 1 Adrenal gland = absolute adrenal gland weight (g)/body weight (kg). Abbreviations: 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control; H/L ratio, heterophil-to-lymphocyte ratio [106].
Table 3. Effects of cecal microbiota transplantation on levels of plasma natural IgG concentrations, and pro- (IL-6 and TNF-α) and anti-inflammatory cytokines (IL-10) of recipient roosters in week 5 and week 16.
Table 3. Effects of cecal microbiota transplantation on levels of plasma natural IgG concentrations, and pro- (IL-6 and TNF-α) and anti-inflammatory cytokines (IL-10) of recipient roosters in week 5 and week 16.
TreatmentIgG
(mg/mL)
IL-6
(pg/mL)
TNF-α
(pg/mL)
IL-10
(pg/mL)
Week 5
CTRL5.19738.53222.84642.569
72-CMT5.41237.10926.49533.259
63-CMT5.24532.90326.21137.503
SEM0.6242.0142.5975.254
p-value0.5650.1180.2930.499
Week 16
CTRL15.032 ab43.128 AB16.66027.467 ab
72-CMT17.993 a47.523 A21.70626.928 b
63-CMT13.716 b38.597 B16.16133.835 a
SEM1.1763.2941.8961.997
p-value0.0460.0700.1070.045
Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05), A,B show trend differences (0.05 < p ≤ 0.10). Abbreviations: 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control; IL, interleukin; TNF-α, tumor necrosis factor alpha [106].
Table 4. Effects of cecal microbiota transplantation on mucosal sIgA concentrations and splenic relative mRNA abundance of pro- (IL-6 and TNF-α) and anti-inflammatory cytokines (IL-10) of recipient roosters in week 5 and week 16.
Table 4. Effects of cecal microbiota transplantation on mucosal sIgA concentrations and splenic relative mRNA abundance of pro- (IL-6 and TNF-α) and anti-inflammatory cytokines (IL-10) of recipient roosters in week 5 and week 16.
TreatmentsIgA
(mg/g)
Relative mRNA Abundance
IL-6TNF-αIL-10
Week 5
CTRL2.167 ab0.8060.9050.396
72-CMT1.757 b0.7631.3780.461
63-CMT3.473 a0.6731.2800.258
SEM0.4400.1410.1750.153
p-value0.0450.7960.2960.456
Week 16
CTRL6.4331.133 AB2.390 AB0.879
72-CMT7.9891.694 A2.741 A0.739
63-CMT9.9140.832 B2.217 B0.816
SEM1.3690.2630.1490.266
p-value0.2490.0800.0650.722
Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05), A,B show trend differences (0.05 < p ≤ 0.10). Abbreviations: 63-CMT, chickens with cecal bacterial solution of donor line 63; 72-CMT, chickens with cecal bacterial solution of donor line 72; CTRL, control; IL, interleukin; sIgA, secretory immunoglobulin A; TNF-α, tumor necrosis factor alpha [106].
Table 5. Effects of cecal microbiota transplantation on MAOA mRNA expression, serotonergic activities, dopamine, and norepinephrine in the hypothalamus of recipient chickens in week 5 and week 16.
Table 5. Effects of cecal microbiota transplantation on MAOA mRNA expression, serotonergic activities, dopamine, and norepinephrine in the hypothalamus of recipient chickens in week 5 and week 16.
TreatmentMAOA5-HT
(ng/g)
5-HIAA
(ng/g)
5-HIAA/5-HTTryptophan
(ng/g)
Dopamine
(ng/g)
Norepinephrine
(ng/g)
Week 5
63-CMT1.51496 a122.3 a0.2251784 A55.7 ab394 AB
72-CMT1.56388 b86.7 b0.2311532 AB44.5 b324 B
CTRL1.45482 ab108.3 ab0.2251454 B70.6 a494 A
SEM0.1129.46.90.014103.66.628.1
p-value0.800.040.0070.5000.090.030.07
Week 16
63-CMT2.52 a39759.30.164 a2760121540
72-CMT1.80 b36850.90.131 ab2140121517
CTRL1.90 ab384420.110 b2480117506
SEM0.0529.55.20.0121769.630.7
p-value0.020.800.230.0110.060.940.71
Values are least-squares means ± SEM, n = 7. a,b indicate significant differences (p ≤ 0.05), A,B indicate trend differences (0.05 < p ≤ 0.10). Abbreviations: 5-HT, serotonin; 5-HAA, 5-hydroxuindoleacetic acid; 63-CMT, received cecal content solution from 63 donors; 72-CMT, received cecal content solution from 72 donors; CTRL, received saline, control; MAOA, monoamine oxidase A [123].
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

Fu, Y.; Hu, J.; Zhang, H.; Erasmus, M.A.; Johnson, T.A.; Cheng, H.-W. The Impact of Early-Life Cecal Microbiota Transplantation on Social Stress and Injurious Behaviors in Egg-Laying Chickens. Microorganisms 2024, 12, 471. https://doi.org/10.3390/microorganisms12030471

AMA Style

Fu Y, Hu J, Zhang H, Erasmus MA, Johnson TA, Cheng H-W. The Impact of Early-Life Cecal Microbiota Transplantation on Social Stress and Injurious Behaviors in Egg-Laying Chickens. Microorganisms. 2024; 12(3):471. https://doi.org/10.3390/microorganisms12030471

Chicago/Turabian Style

Fu, Yuechi, Jiaying Hu, Huanmin Zhang, Marisa A. Erasmus, Timothy A. Johnson, and Heng-Wei Cheng. 2024. "The Impact of Early-Life Cecal Microbiota Transplantation on Social Stress and Injurious Behaviors in Egg-Laying Chickens" Microorganisms 12, no. 3: 471. https://doi.org/10.3390/microorganisms12030471

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