You are on page 1of 8

Microbial endocrinology: Why the intersection of microbiology and

neurobiology matters to poultry health

Daniel N. Villageliũ and Mark Lyte1

Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State
University, 1800 Christensen Drive, Ames, IA 50011–1134

ABSTRACT The union of microbiology and neurobi- physiological functions. The realization that the gut mi-
ology has led to a revolution in the way we view the mi- crobiome serves as a kind of “microbial organ” has im-
crobiome. Now recognized as important symbionts, the portant implications for many areas of biology. In this
microorganisms which inhabit multiple niches in mam- paper advances in the field of microbial endocrinology
malian and avian (chicken) hosts, such as the intesti- which may hold relevance for the poultry industry are
nal tract and skin, serve and influence many important examined.
Key words: microbiome, neurobiology, enteric nervous system, stress, behavior
2017 Poultry Science 96:1–8
http://dx.doi.org/10.3382/ps/pex148

INTRODUCTION and was used in the treatment of urticaria. (Commonly


referred to as hives, urticaria is a condition classically
Microbial endocrinology represents the union of the described as raised and itchy areas of the skin which
fields of microbiology and neurobiology. It is defined may indicate exposure to an allergen). Reports of pa-
as the study of the ability of microorganisms to pro- tients suffering from fulminating sepsis mere hours af-
duce and recognize neurochemicals that originate ei- ter treatment with epinephrine began to appear. The
ther within the microorganisms themselves or within cause was traced back to syringes used to drain bac-
the host they inhabit. While term originated in 1993 terial abscesses. Though abscess drainage was a com-
(Lyte, 1993), evidence supporting some of the principle mon surgical practice and practitioners of the day were
tenants of this field can be traced to studies going back likely vigilant in the use of cleaning agents, methods
as far as 1930. For a poultry producer two issues stand of the time were unable to completely sterilize spore
out as especially important: forming organisms like C. perfringens. Ordinarily, the
remaining spores were generally dormant and appar-
(1) Microorganisms do not live in isolation from each ently below the threshold necessary to cause infection.
other or their host. They communicate with, in- However, with exposure to epinephrine, the organism’s
fluence, and are influenced by their host and their infective potential changes. Epinephrine increases the
neighboring microbes. growth rate of C. perfringens and decreases the in-
(2) Systems of host-microbe communication appear fective dose by up to a million-fold (Renaud, 1930;
across a wide range of organisms from plants to Cooper, 1946). Cases related to this phenomenon were
animals. reported throughout the 20th century. In 1968 Harvey
and Purnell commented on a fatal case of gas gangrene
in a 22-year-old who received intramuscular epinephrine
Microorganisms actively respond to signals produced
(Harvey and Purnell, 1968). Similarly, in 1983 a clinical
by the host in very significant ways. For example, the
report described the development of gas gangrene in a
same agent responsible for multibillion dollar losses
13-year-old girl (Teo and Balasubramaniam, 1983).
through necrotic enteritis in chickens, Clostridium per-
The potential for neurotransmitters to influence vir-
fringens, also turns out to be among the earliest docu-
ulence is not solely limited to Clostridium. The growth
mented cases of bacterial growth being influenced by a
of the respiratory pathogen Bordetella bronchiseptica
stress related neurochemical.
is greatly increased by the presence of norepinephrine
In the early 1930 s, epinephrine was the first man-
in vitro (Anderson and Armstrong, 2008). The release
ufactured hormone to be used in the clinical setting
of catecholamines from injured enteric neurons is asso-
ciated with a rapid alteration of the microbiota com-

C 2017 Poultry Science Association Inc.
munity from one dominated by Gram-positive taxa
Received April 3, 2017.
Accepted May 9, 2017. to one dominated by a single Gram-negative bacte-
1
Corresponding author: mlyte@iastate.edu rial species, Escherichia coli (Lyte and Bailey, 1997).

1
2 Villageliũ and Lyte

The overgrowth of E. coli has been proposed to be a duced by the microbiome influence a range of physi-
possible contributory factor in trauma-induced sepsis ological and behavioral functions extending from main-
(Freestone et al., 2002). Findings like this may hold tenance of the gastrointestinal (GI) tract to iron uptake
useful implications. For example, if the effects of stress by bacteria (Gross and Siegel, 1988; Freestone et al.,
hormones could be suppressed, outbreaks of necrotic 2000; Grenham et al., 2011).
enteritis in chickens might also be diminished. The con- In addition to infectious considerations, it should also
cept is not necessarily far off. Dopamine antagonists are be appreciated that microbial endocrinology has rel-
chemicals which block dopamine receptors through an- evance outside of the field of infectious disease. Neu-
tagonistic binding of the receptor. Often used in the rochemicals, isolated from microorganisms, which have
treatment of psychiatric illnesses such as schizophre- been shown to have biological activity in mammalian
nia, dopaminergic antagonists have been demonstrated cells include corticotropin from Tetrahymena pyri-
to block catecholamine-induced growth in Escherichia formis, somatostatin from Bacillus subtilis, and pro-
coli O157: H7, Salmonella enterica, and Yersinia ente- gesterone from Trichophyton mentagrophytes (LeRoith
rocolitica (Freestone et al., 2007). et al., 1985; Schar et al., 1986). When viewed from
Though neurochemical influences on the growth the standpoint of the microorganism, it becomes ap-
rate of pathogens are of obvious importance to in- parent that the functions of many of these chemicals
fectious disease, it should be noted that the mech- are not pathological in nature. To successfully develop
anisms through which neurochemicals achieve physi- the strong and reliable approaches needed to modify in
ological changes may be quite varied. Consider the vivo systems, a more evolutionary-based approach must
respiratory pathogen Actinobacillus pleuropneumoniae be utilized to explore the mechanisms through which
which shows differential expression of 158 and 105 genes microorganisms can influence their host. This includes
respectively, for organisms exposed to epinephrine viewing these neurochemicals from the point of view of
or norepinephrine. Though both chemicals are cate- the microorganism as well as the host.
cholamines, only 18 of the differentially expressed genes
were common to both epinephrine and norepinephrine THE MICROBIOTA-GUT-BRAIN AXIS AND
exposure. This suggests that organisms like A. pleu-
ropneumoniae may possess multiple responsive sys- SUPPORT FOR ITS INCLUSION IN AN
tems for individual catecholamines (Li et al., 2012). AVIAN MODEL
This is not surprising; there are several distinct classes In mammalian models, one physiological mechanism
of catecholamine receptors ranging from dopaminergic by which microbes interact with their host has been
to adrenergic, each with several subtypes and an un- termed the microbiota-gut-brain axis (Figure 1). Food
counted numbers of heteromers. The range of neuro- ingested by the host can be metabolized by the mi-
chemicals and the variety of microorganisms in which crobiota in ways that generate or release neuroactive
they have been identified is quite extensive (Lenard, components. These neuroactive molecules can be taken
1992). Examples include acetylcholine (Stephenson and up into the circulation or interact with the host locally
Rowatt, 1947; Kawashima et al., 2007), histamine at the level of the enteric nervous system. Some signals
(Devalia, 1988; Masson et al., 1996; Thomas et al., may be transduced to the brain where they can influ-
2012), serotonin (Hurley et al., 1971; Ozogul, 2011), ence cognition and behavior whereas other signals may
catecholamines (Tsavkelova et al., 2000; Shishov et al., result in changes in the enteric system which feedback
2009; Asano et al., 2012), and agmatine (Raasch et al., on the microbiota (Jarvis, 2005; Powley et al., 2011;
1995; Arena and Manca de Nadra, 2001). Furness et al., 2014).
Given the observations above, it is also not surpris-
ing to learn that physiological changes caused by neu-
rotransmitters are similarly quite varied. For example, Is there evidence for the modulation of
it has been demonstrated that concentrations of nore- chicken behavior through a
pinephrine as low as 1.2 × 10−5 M could significantly microbiota-gut-brain axis?
influence the aerotolerance of organisms like Clostrid-
ium jejuni and Spirillum volutsans (Bowdre et al., Recent advances in neurobiology have shown a great
1976). At concentrations this low, norepinephrine is deal of similarity between avian neural structure and
not a useful metabolite but it can still serve as a sig- that of their mammalian cousins. It has been demon-
naling molecule. Generally, C. jejuni and S. volutsans strated that the avian brain has cortex-like struc-
can only be cultured in a microaerophilic environment. tures which are not unlike the structures found in a
In the presence of catecholamines however, these or- mammalian brain (Jarvis, 2005). The avian pallium
ganisms become more aerotolerant. If norepinephrine contains functional columns similar to those in the
is present, Spirillum inoculations can successfully be mammalian cortex. The cortex is the outer layer of cere-
established in oxygen containing environments using bral gray matter associated with what can be crudely
inoculums smaller than what was necessary without described as “higher brain function” which includes,
catecholamines (Bowdre et al., 1976). Over the years but is not limited to, activities such as coordination of
authors have suggested that neurotransmitters pro- sensory information, voluntary movement, and learning
Relevance of microbial endocrinology to poultry 3

Figure 1. A hypothetical gut-brain axis in chickens.

and memory. Structures analogous to the mammalian STRESS MANAGEMENT AND OTHER
septal neuroendocrine system can also be found in avian POTENTIAL APPLICATIONS OF
species (Kuenzel, 2014). Septal nuclei are part of the
MICROBIAL ENDOCRINOLOGY FOR THE
limbic system which is a region of the brain associated
with motivation and emotion. The limbic system can POULTRY INDUSTRY
exert influence through the endocrine system as well as In reviewing the state of the poultry industry, at least
autonomic system. six major issues become quickly apparent. Feed cost,
Similarities between the chicken gut microbiome disease outbreaks, discontinuation of feed antibiotics,
and mammalian counterparts are also apparent. The poultry welfare, and issues related to the environment.
avian small intestine harbors large populations of Although the solutions to these complex problems will
Lactobacillus and Enterococcus. Common phyla found likely be multifactorial, approaches to all of these issues
in the chicken GI tract are Firmicutes, Bacteroides, can be conceived of with the aid of microbial endocrinol-
Proteobacteria, and Actinobacteria (Oakley et al., ogy.
2014). This is not unlike reports of the mammalian Stress in chickens can lead to numerous issues rang-
microbiome being dominated by Firmicutes (65.7% ing from cannibalism to the premature termination of
by sequencing), Bacteroidetes (16.3%), Proteobacteria egg production. Stressful factors for a chicken are some-
(8.8%), and Actinobacteria (4.7%) (Ley et al., 2008). times surprising and difficult to predict. Even the move
Given the abundance of similarities between birds to less restrictive environments can result in stresses
and mammals, it seems highly probable that an avian that can cause a profound impact. For example, a re-
gut-brain axis should exist. Until recently, evidence sup- cent study found that mortality was over twice as high
porting this contention was elusive. However, reports in less restricted aviary reared hens (5.52%) in compar-
in chickens is now available which shows a relation- ison to cage reared hens (2.48%) during their producing
ship between intestinal inflammation, behavioral pat- phase (Tahamtani et al., 2014). A large proportion of
terns, corticosterone serum levels, and central nervous the birds that were found dead had bloody sores to the
system activity (Calefi et al., 2016). Chickens exposed head and neck region indicative of injurious pecking.
to intestinal stressors were found to have activation of The development of injurious pecking behavior in ex-
the hypothalamic-pituitary-adrenal axis as well as in- tended range chickens highlights the potential for the
creased corticosterone serum levels. Interestingly, ani- environment to exacerbate social stresses in unforeseen
mals infected with Clostridium perfringens and exposed ways.
to other endocrine influencing conditions such as heat How could microbial endocrinology play a role in the
demonstrated fewer symptomatic behaviors and less management of stress? The link between social stress
intestinal lesions than animals solely challenged with and the neuroendocrine system is well established if not
Clostridium perfringens. Albeit correlational, this work completely understood. Social stress can act through
provides useful insight and validates research efforts the central nervous system to trigger the release of neu-
which seek to improve livestock productivity through rotransmitters and stress hormones from the sympa-
influences on neuroendocrine pathways. thoadrenal and hypothalamic-pituitary-adrenal axes. In
4 Villageliũ and Lyte

Figure 2. Social stress activates the central nervous system. (1) The stress signal may be transduced by neurons of the autonomic nervous
system as well as the hypothalamic-pituitary-adrenal (HPA) axis. (2) Chemicals released by these systems can act on the microbiome. (3) Changes
in host microbiome and the production of metabolites may then be detected by the host through the gut-brain axis (4) and transmitted by vagal
sensory neurons back to the central nervous system (CNS) or otherwise effect changes in the host (5).

chickens, physiological differences have been observed the microbiome and host are intimately linked in a bi-
between behaviorally distinct dominant and subordi- directional way (Figure 3).
nate animals. Such differences include differing levels Evidence supporting this model is abundant. Numer-
of dopamine and dopamine receptors in the dopaminer- ous studies have demonstrated that stress can affect
gic system as well as hypertrophy of the adrenal glands the gut’s microbial composition, influence microbiota-
and changes in corticosteroid levels in the adrenergic gut-brain communication, and result in behavioral
system (Harvey et al., 1984; Blanchard et al., 1993; alteration (Cryan and Dinan, 2012; Collins et al.,
Shively, 1998; Fano et al., 2001). Some have suggested 2013; David et al., 2014). Physical and psychosocial
that changes such as these may result in abnormal and stress, as well as alteration of the circadian rhythm,
detrimental behaviors or an inability for chickens to can alter the microbial community structure within
cope with environmental stresses (Cheng et al., 2002). the gut (Bailey et al., 2011; Bangsgaard Bendtsen
Such outcomes are far from benign. It has been reported et al., 2012; Thaiss et al., 2014). The microbial com-
that chickens with a poor ability to adjust to their social munity structure can rapidly change as a response
environment also display a greater susceptibility to dis- to the influx of host stress-related neurochemicals in
ease (Gross and Siegel, 1988; Awadalla, 1998). Behav- the lumen.
ioral consequences can include an increased tendency The implications of this model for chickens are as
towards cannibalism, aggression, and feather pecking of yet mostly unexplored. Stress related disruptions to
(Burger and Kaiser, 1996; Via, 1999; Bilčı́k and Keel- the chicken microbiome could conceivably affect multi-
ing, 2000; El-Lethey et al., 2000). ple systems in deleterious ways. However, we can in-
Using an understanding of microbial endocrinology, fer from recent research that susceptibility to infec-
we can re-evaluate these host-centric models of social tion would be one area of great interest. In rodents,
stress to expand on the ideas above. As shown in Fig- investigations have shown that social conflict stress
ure 1, the concept of animal social stress should in- leaves the animal more susceptible to infection. In
corporate the dynamic role that the microbiome plays 1991 to 1992 a series of experiments by Lyte et al.
in responding to stress (Figure 2). Briefly, social stress were conducted with animals stressed by social conflict
acts through the central nervous system and effects (Foster et al., 2016; Lyte, 2016). These stressed animals
changes to the microbiome through the sympathoad- were then challenged with the oral pathogen Yersinia
renal and hypothalamic-pituitary-adrenal axes. These enterocolitica. After 14 days, the stressed population
neurochemical axes release chemicals which can act on showed over 100% greater mortality. Cumulative sur-
and change the microbiome. Changes in the host’s mi- vival rates of 80% for non-stressed infected animals but
crobiome may then feedback onto the host through the less than 40% survival of stressed animals. Crucially,
gut-brain axis to effect further changes. Simply stated, Lyte went on to show a mechanistic link between stress
Relevance of microbial endocrinology to poultry 5

Figure 3. Bi-directional nature of microbial endocrinology in which neurochemicals produced by the host can influence the microbiota (A) and
the very same neurochemicals produced by the microbiota can influence the host (B). The evolutionary-based neurochemical signaling pathway
between microbiota and host means that a neurochemical(s) produced by the host can influence the microbiota (A) and at the same time a
neurochemical(s) produced by the microbiota can, in turn, influence the host (B).

Figure 4. Initial experiment in microbial endocrinology. Yersinia enterocolitica culture plates in 1991 showing that bacterial growth in serum-
based medium was enhanced in the presence of the neuroendocrine stress hormone norepinephrine, but not epinephrine or control diluent (Lyte,
1993; Lyte, 2010).

and the behavior of the microbes (Lyte, 1993). Cate- poultry. Though Campylobacter often asymptomati-
cholamines, the key chemical modulators of stress re- cally colonizes the chicken GI track, sometimes the
sponses also induce growth changes in Y. enterocolit- species is associated with hemorrhagic enteritis, focal
ica. Those catecholamines which are found in the GI hepatic necrosis, and chick death. What determines
tract and are released as a consequence of stress, such whether an organism behaves in a benign or invasive
as dopamine and norepinephrine, increased the growth often depends on how the organism perceives its en-
of Y. enterocolitica over a million-fold. Y. enterocolit- vironment. Perhaps direct neurochemical-bacterial in-
ica, which is almost exclusively a gut pathogen, does teractions as already observed (and discussed above)
not respond to epinephrine (Figure 4). This is likely be- for several Gram-negative bacteria can be further ex-
cause the enzyme responsible for producing epinephrine tended to explain the differential infective behavior of
from norepinephrine is not present in the enteric ner- Campylobacter in chickens? If pathogenic microorgan-
vous system. Thus, Y. enterocolitica which has adapted isms perceive a stressed environment, this can lead to
to a gastrointestinal environment in which epinephrine increased pathogen colonization which in turn can lead
is not present has no reason to detect or respond to more highly contaminated food and a greater risk of
to epinephrine. foodborne infection.
The results of this study may be generalized to poul-
try science. While the poultry producer may have lit-
tle concern with Y. enterocolitica, the stress neuro- Alteration of the microbiome
chemical norepinephrine has also been shown to stim- As the role of the microbiome in health and disease
ulate the growth and pathogenicity of other organ- is increasingly recognized, it follows that efforts to op-
isms including E. coli and Campylobacter jejuni (Aroori timize the health of the organism would be better in-
et al., 2014). C. jejuni is the predominant species as- formed with a more complete understanding of the mi-
sociated with human foodborne infection derived from crobiome. It may be questioned, what should an ideal
6 Villageliũ and Lyte

microbiome look like and how could one go about in- CONCLUDING REMARKS
fluencing or optimizing it? This question is of active
interest with many publications still offering differing The influence of the microbiome on host physiol-
perspectives. However, while research in this field is ogy is an area of increasing poultry research especially
still in its infancy with correlational, not mechanistic due to the introduction of new generation molecular
studies pre-dominating the literature, some useful ob- techniques that are revolutionizing the field of microbi-
servations have been made. These observations include ology itself. It is proposed that observations and re-
(but are not limited to) the following: search undertaken from the perspective of microbial
endocrinology could lead to useful applications. For ex-
(1) Rich microbial diversity appears to be associated ample, could the design of new dietary approaches to
with better host health whereas decreased diversity colonize the chicken GI tract with microorganisms ca-
has been associated with health problems rang- pable of inactivation of stress-induced catecholamine re-
ing from recurrent Clostridium difficile infection lease in the gut lead to increased resilience to pathogen
to metabolic disturbances (Chang et al., 2008; colonization in stressed poultry? Are there other as yet
Claesson et al., 2012). unrealized benefits to poultry health that may be re-
(2) Diet is one of the most important modifiers of the alized by selecting for specific microbiomes based on
intestinal microbiome and can influence its compo- microbial endocrinology-based approaches? Ultimately,
sition and functional metabolism (Albenberg and the benefits that can be realistically obtained can only
Wu, 2014; David et. al. 2014). This is not limited to be determined with more research. Through a rigorous
digestible material. Dietary components like fiber, study of the microbial endocrinology-based mechanisms
xenobiotics, neurotransmitters, and their precur- by which microbiomes influence their host, we may de-
sors can all have an influence. For example, Suez velop tools that will help address the problems facing
et al. found that artificial sweeteners can enrich for the chicken industry today.
Bacteroides (Suez et al., 2014).

As was discussed earlier, the behavior of a mi-


REFERENCES
croorganism often depends on how the organism per- Albenberg, L. G., and G. D. Wu. 2014. Diet and the intestinal mi-
ceives its environment. Obviously, a great deal of a crobiome: Associations, functions, and implications for health and
disease. Gastroenterology 146:1564–1572.
gut microbe’s environment will depend on the diet Anderson, M. T., and S. K. Armstrong. 2008. Norepinephrine medi-
of the host organism. It has been established that ates acquisition of transferrin-iron in bordetella bronchiseptica. J
the diet can profoundly impact the levels of certain Bacteriol 190:3940–3947.
neurochemicals in a host (Fernstrom and Fernstrom, Arena, M. E., and M. C. Manca de Nadra. 2001. Biogenic amine
production by lactobacillus. J Appl Microbiol 90:158–162.
2007) demonstrated that the rates of synthesis for sero- Aroori, S. V., T. A. Cogan, and T. J. Humphrey. 2014. Effect of no-
tonin and catecholamines in the brain are sensitive radrenaline on the virulence properties of campylobacter species.
to local substrate concentrations. Physiologic factors Int J Microbiol 2014:279075.
(particularly diet) which influence the pool of neu- Asano, Y., T. Hiramoto, R. Nishino, Y. Aiba, T. Kimura, K. Yoshi-
hara, Y. Koga, and N. Sudo. 2012. Critical role of gut microbiota
rochemical precursors in the brain also influence the in the production of biologically active, free catecholamines in
synthetic rate of neurochemical products. This finding the gut lumen of mice. Am J Physiol Gastrointest Liver Physiol
has functional consequences. For example, dietary con- 303:G1288–1295.
sumption of tyrosine, the catecholamine precursor, in- Awadalla, S. F. 1998. Effect of some stressors on pathogenicity of
eimeria tenella in broiler chicken. J Egypt Soc Parasitol 28:683–
creases anger in subjects exposed to psychological stress 690.
(Lieberman, 2015). It is of note that corn is the grain Bailey, M. T., S. E. Dowd, J. D. Galley, A. R. Hufnagle, R. G.
most routinely used in commercial poultry diets in the Allen, and M. Lyte. 2011. Exposure to a social stressor alters the
United States. Interestingly, a reduced level of brain structure of the intestinal microbiota: Implications for stressor-
induced immunomodulation. Brain Behav Immun 25:397–407.
serotonin and an elevated level of catecholamines can be Bangsgaard Bendtsen, K. M., L. Krych, D. B. Sorensen, W. Pang, D.
found in animals fed only a tryptophan-poor tyrosine- S. Nielsen, K. Josefsen, L. H. Hansen, S. J. Sorensen, and A. K.
rich diet such as corn (Fernstrom, 1977). Hansen. 2012. Gut microbiota composition is correlated to grid
floor induced stress and behavior in the balb/c mouse. PLoS One
Just as a host organism can synthesize more neuro- 7:e46231.
transmitters with an abundance of precursors, so can Bilčı́k, B., and L. J. Keeling. 2000. Relationship between feather
microbes. Microbes which produce neurotransmitters pecking and ground pecking in laying hens and the effect of group
often use the same conserved synthetic pathways as size. Applied Animal Behaviour Science 68:55–66.
Blanchard, D. C., R. R. Sakai, B. McEwen, S. M. Weiss, and R.
their eukaryotic host (Lyte, 2014). Therefore, it would J. Blanchard. 1993. Subordination stress: Behavioral, brain, and
not be surprising to learn that increased dietary con- neuroendocrine correlates. Behav Brain Res 58:113–121.
sumption of food substrates which are neurochemical Bowdre, J. H., N. R. Krieg, P. S. Hoffman, and R. M. Smibert. 1976.
precursors, could also lead to an abundance of neu- Stimulatory effect of dihydroxyphenyl compounds on the aerotol-
erance of spirillum volutans and campylobacter fetus subspecies
rochemicals in the gut. Once produced, these neuro- jejuni. Appl Environ Microbiol 31:127–133.
transmitters could influence the microbiome and the Burger, H., and H. E. Kaiser. 1996. Crowding. In Vivo 10:249–253.
host which would in turn influence each other bi- Calefi, A. S., J. G. da Silva Fonseca, D. W. Cohn, B. T. Honda, C.
directionally (Figure 3). Costola-de-Souza, L. E. Tsugiyama, W. M. Quinteiro-Filho, A.
Relevance of microbial endocrinology to poultry 7
J. Piantino Ferreira, and J. Palermo-Neto. 2016. The gut-brain Harvey, P. W., and G. V. Purnell. 1968. Fatal case of gas gangrene
axis interactions during heat stress and avian necrotic enteritis. associated with intramuscular injections. Br Med J 1:744–746.
Poult Sci 95:1005–1014. Harvey, S., J. G. Phillips, A. Rees, and T. R. Hall. 1984. Stress and
Chang, J. Y., D. A. Antonopoulos, A. Kalra, A. Tonelli, W. T. Khal- adrenal function. J Exp Zool 232:633–645.
ife, T. M. Schmidt, and V. B. Young. 2008. Decreased diversity of Hurley, R., B. G. Leask, C. R. Ruthven, M. Sandler, and J. South-
the fecal microbiome in recurrent clostridium difficile-associated gate. 1971. Investigation of 5-hydroxytryptamine production by
diarrhea. J Infect Dis 197:435–438. candida albicans in vitro and in vivo. Microbios 4:133–143.
Cheng, H. W., P. Singleton, and W. M. Muir. 2002. Social stress in Jarvis, E. D., Gunturkun, L. Bruce, A Csillag, H. Karten, W.
laying hens: Differential dopamine and corticosterone responses Kuenzel, L. Medina, G. Paxinos, D. J. Perkel, T. Shimizu, G.
after intermingling different genetic strains of chickens. Poult Sci Streiedter, J. M. Wild, G. F. Ball, J. Dugas-Ford, S. E. Durand,
81:1265–1272. G. E. Hough, S. Husband, L. Kubikova, D. W. Lee, C. V. Mell, A.
Claesson, M. J., I. B. Jeffery, S. Conde, S. E. Power, E. M. O’Connor, Powers, C. Siang, T. V. Smulders, K. Wada, S. A. White, K. Ya-
S. Cusack, H. M. Harris, M. Coakley, B. Lakshminarayanan, O. mamoto, J. Yu, A. Reiner, and A. B. Butler. 2005. Avian brains
O’Sullivan, G. F. Fitzgerald, J. Deane, M. O’Connor, N. Harnedy, and a new understanding of vertebrate brain evolution. Nat Rev
K. O’Connor, D. O’Mahony, D. van Sinderen, M. Wallace, L. Neurosci 6:151–159.
Brennan, C. Stanton, J. R. Marchesi, A. P. Fitzgerald, F. Shana- Kawashima, K., H. Misawa, Y. Moriwaki, Y. X. Fujii, T. Fujii, Y.
han, C. Hill, R. P. Ross, and P. W. O’Toole. 2012. Gut microbiota Horiuchi, T. Yamada, T. Imanaka, and M. Kamekura. 2007. Ubiq-
composition correlates with diet and health in the elderly. Nature uitous expression of acetylcholine and its biological functions in
488:178–184. life forms without nervous systems. Life Sci 80:2206–2209.
Collins, S. M., Z. Kassam, and P. Bercik. 2013. The adoptive trans- Kuenzel, W. J. 2014. Research advances made in the avian brain and
fer of behavioral phenotype via the intestinal microbiota: Exper- their relevance to poultry scientists. Poult Sci 93:2945–2952.
imental evidence and clinical implications. Curr Opin Microbiol Lenard, J. 1992. Mammalian hormones in microbial cells. Trends
16:240–245. Biochem Sci 17:147–150.
Cooper, E. V. 1946. Gas-gangrene following injection of adrenaline. LeRoith, D., W. Pickens, A. I. Vinik, and J. Shiloach. 1985. Bacil-
Lancet 1:459–461. lus subtilis contains multiple forms of somatostatin-like material.
Cryan, J. F., and T. G. Dinan. 2012. Mind-altering microorganisms: Biochem Biophys Res Commun 127:713–719.
The impact of the gut microbiota on brain and behaviour. Nat Ley, R. E., M. Hamady, C. Lozupone, P. Turnbaugh, R. R. Ramey,
Rev Neurosci 13:701–712. J. S. Bircher, M. L. Schlegel, T. A. Tucker, M. D. Schrenzel, R.
David, L. A., C. F. Maurice, R. N. Carmody, D. B. Gootenberg, J. Knight, and J. I. Gordon. 2008. Evolution of mammals and their
E. Button, B. E. Wolfe, A. V. Ling, A. S. Devlin, Y. Varma, gut microbes. Science 320:1647–1651.
M. A. Fischbach, S. B. Biddinger, R. J. Dutton, and P. J. Li, L., Z. Xu, Y. Zhou, L. Sun, Z. Liu, H. Chen, and R. Zhou. 2012.
Turnbaugh. 2014. Diet rapidly and reproducibly alters the human Global effects of catecholamines on actinobacillus pleuropneumo-
gut microbiome. Nature 505:559–563. niae gene expression. PLoS One 7:e31121.
Devalia, JL, Y. H., DR Cundell, RJ Davies, D Grady, and S. Lieberman, H. R., L. A. Thompson, C. M. Caruso, P. J. Niro, C.
Tabaqchali 1988. Variation in histamine synthesis by gram- R. Mahoney, J. P. McClung, and G. R. Caron 2015. The cat-
negative and gram-positive respiratory tract bacteria and the ef- echolamine neurotransmitter precursor tyrosine increases anger
fect of cefaclor. Royal Society of Medicine Services Ltd Interna- during exposure to severe psychological stress. Psychopharma-
tional Congress and Symposium Series. 49–55. cology 232:943–951.
El-Lethey, H., V. Aerni, T. W. Jungi, and B. Wechsler. 2000. Stress Lyte, M. 1993. The role of microbial endocrinology in infectious dis-
and feather pecking in laying hens in relation to housing condi- ease. J Endocrinol 137:343–345.
tions. Br Poult Sci 41:22–28. Lyte, M. 2010. Microbial endocrinology: A personal journey. Pages
Fano, E., J. R. Sanchez-Martin, A. Arregi, B. Castro, A. Alonso, P. 1–16 In Microbial endocrinology: Interkingdom signaling in in-
Brain, and A. Azpiroz. 2001. Social stress paradigms in male mice: fectious disease and health. M. Lyte, and P. E. P Freestone eds.
Variations in behavior, stress and immunology. Physiol Behav Springer New York, New York, NY.
73:165–173. Lyte, M. 2014. Microbial endocrinology and the microbiota-gut-
Fernstrom, J. D. 1977. Effects on the diet on brain neurotransmit- brain axis. Adv Exp Med Biol 817:3–24.
ters. Metabolism 26:207–223. Lyte, M. 2016. Microbial endocrinology: An ongoing personal jour-
Fernstrom, J. D., and M. H. Fernstrom. 2007. Tyrosine, phenylala- ney. Adv Exp Med Biol 874:1–24.
nine, and catecholamine synthesis and function in the brain. J Lyte, M., and M. T. Bailey. 1997. Neuroendocrine-bacterial inter-
Nutr 137:1539S–1547S; discussion 1548S. actions in a neurotoxin-induced model of trauma. J Surg Res
Foster, J. A., M. Lyte, E. Meyer, and J. F. Cryan. 2016. Gut micro- 70:195–201.
biota and brain function: An evolving field in neuroscience. Int J Masson, F., R. Talon, and M. C. Montel. 1996. Histamine and tyra-
Neuropsychopharmacol 19:1–7. mine production by bacteria from meat products. Int J Food Mi-
Freestone, P. P., R. D. Haigh, and M. Lyte. 2007. Blockade of crobiol 32:199–207.
catecholamine-induced growth by adrenergic and dopaminergic Oakley, B. B., H. S. Lillehoj, M. H. Kogut, W. K. Kim, J. J. Mau-
receptor antagonists in escherichia coli o157:H7, salmonella en- rer, A. Pedroso, M. D. Lee, S. R. Collett, T. J. Johnson, and N.
terica and yersinia enterocolitica. BMC Microbiology 7:1–13. A. Cox. 2014. The chicken gastrointestinal microbiome. FEMS
Freestone, P. P., P. H. Williams, R. D. Haigh, A. F. Maggs, Microbiol Lett 360:100–112.
C. P. Neal, and M. Lyte. 2002. Growth stimulation of in- Ozogul, F. 2011. Effects of specific lactic acid bacteria species on
testinal commensal escherichia coli by catecholamines: A pos- biogenic amine production by foodborne pathogens. Int J Food
sible contributory factor in trauma-induced sepsis. Shock Sci Technol:478–484.
18:465–470. Powley, T. L., R. A. Spaulding, and S. A. Haglof. 2011. Vagal af-
Freestone, P. P. E., M. Lyte, C. P. Neal, A. F. Maggs, R. D. Haigh, ferent innervation of the proximal gastrointestinal tract mucosa:
and P. H. Williams. 2000. The mammalian neuroendocrine hor- Chemoreceptor and mechanoreceptor architecture. J Comp Neu-
mone norepinephrine supplies iron for bacterial growth in the rol 519:644–660.
presence of transferrin or lactoferrin. J Bacteriol 182:6091–6098. Raasch, W., S. Regunathan, G. Li, and D. J. Reis. 1995. Agmatine,
Furness, J. B., B. P. Callaghan, L. R. Rivera, and H. J. Cho. 2014. the bacterial amine, is widely distributed in mammalian tissues.
The enteric nervous system and gastrointestinal innervation: In- Life Sci 56:2319–2330.
tegrated local and central control. Adv Exp Med Biol 817:39–71. Renaud, M. A. 1930. Role favorisant des perturbations locales causes
Grenham, S., G. Clarke, J. F. Cryan, and T. G. Dinan. 2011. Brain– par l’ adrenaline sur led developpement des infections microbi-
gut–microbe communication in health and disease. Front Physiol ennes. C R Seances Soc Biol Fil 103:1052–1054.
2:94. Schar, G., E. P. Stover, K. V. Clemons, D. Feldman, and D. A.
Gross, W. B., and P. B. Siegel. 1988. Environment-genetic influences Stevens. 1986. Progesterone binding and inhibition of growth in
on immunocompetence. J Anim Sci 66:2091–2094. trichophyton mentagrophytes. Infect Immun 52:763–767.
8 Villageliũ and Lyte

Shishov, VA, T. K., VS Kudrin, and AV. Oleskin . 2009. Amine Teo, W. S., and P. Balasubramaniam. 1983. Gas gangrene after intra-
neuromediators, their precursors, and oxidation products in the muscular injection of adrenaline. Clin Orthop Relat Res:206–207.
culture of escherichia coli k-12. Prikl Biokhim Mikrobiol:550–554. Thaiss, C. A., D. Zeevi, M. Levy, G. Zilberman-Schapira, J. Suez,
Shively, C. A. 1998. Social subordination stress, behavior, and cen- A. C. Tengeler, L. Abramson, M. N. Katz, T. Korem, N. Zmora,
tral monoaminergic function in female cynomolgus monkeys. Biol Y. Kuperman, I. Biton, S. Gilad, A. Harmelin, H. Shapiro, Z.
Psychiatry 44:882–891. Halpern, E. Segal, and E. Elinav. 2014. Transkingdom control of
Stephenson, M., and E. Rowatt. 1947. The production of acetyl- microbiota diurnal oscillations promotes metabolic homeostasis.
choline by a strain of lactobacillus plantarum. J Gen Microbiol Cell 159:514–529.
1:279–298. Thomas, C. M., T. Hong, J. P. van Pijkeren, P. Hemarajata, D. V.
Suez, J., T. Korem, D. Zeevi, G. Zilberman-Schapira, C. A. Trinh, W. Hu, R. A. Britton, M. Kalkum, and J. Versalovic. 2012.
Thaiss, O. Maza, D. Israeli, N. Zmora, S. Gilad, A. Weinberger, Histamine derived from probiotic lactobacillus reuteri suppresses
Y. Kuperman, A. Harmelin, I. Kolodkin-Gal, H. Shapiro, Z. tnf via modulation of pka and erk signaling. PLoS One 7:e31951.
Halpern, E. Segal, and E. Elinav. 2014. Artificial sweeteners in- Tsavkelova, E. A., I. V. Botvinko, V. S. Kudrin, and A. V. Oleskin.
duce glucose intolerance by altering the gut microbiota. Nature 2000. Detection of neurotransmitter amines in microorganisms
514:181–186. with the use of high-performance liquid chromatography. Dokl
Tahamtani, F. M., T. B. Hansen, R. Orritt, C. Nicol, R. O. Moe, Biochem 372:115–117.
and A. M. Janczak. 2014. Does rearing laying hens in aviaries Via, S. 1999. Cannibalism facilitates the use of a novel environment
adversely affect long-term welfare following transfer to furnished in the flour beetle, tribolium castaneum. Heredity (Edinb) 82(Pt
cages? PLoS One 9:e107357. 3):267–275.

You might also like